PMID-sentid Pub_year Sent_text compound_name comp_offset prot_official_name organism prot_offset 33970400-0 2021 The Role of p53 Expression in Patients with RAS/BRAF Wild-Type Metastatic Colorectal Cancer Receiving Irinotecan and Cetuximab as Later Line Treatment. Irinotecan 102-112 tumor protein p53 Homo sapiens 12-15 33930649-11 2021 The green tea extract and EGCG also had a dose-dependent inhibitory effect on irinotecan-induced secretion of interleukin-6 and interleukin-8 by oral epithelial cells. Irinotecan 78-88 interleukin 6 Homo sapiens 110-123 33930649-11 2021 The green tea extract and EGCG also had a dose-dependent inhibitory effect on irinotecan-induced secretion of interleukin-6 and interleukin-8 by oral epithelial cells. Irinotecan 78-88 C-X-C motif chemokine ligand 8 Homo sapiens 128-141 34037277-0 2021 SENP1 participates in Irinotecan resistance in human colon cancer cells. Irinotecan 22-32 SUMO specific peptidase 1 Homo sapiens 0-5 34037277-7 2021 Knockdown of SENP1 reduced the migration ability and trigged re-sensitivity of LoVoR-CPT-11 cells to CPT-11 treatment. Irinotecan 85-91 SUMO specific peptidase 1 Homo sapiens 13-18 34037277-9 2021 In conclusion, SENP1 might play an important role in CPT-11 resistance in colorectal cancer. Irinotecan 53-59 SUMO specific peptidase 1 Homo sapiens 15-20 32743640-0 2021 Irinotecan and vandetanib create synergies for treatment of pancreatic cancer patients with concomitant TP53 and KRAS mutations. Irinotecan 0-10 tumor protein p53 Homo sapiens 104-108 32743640-0 2021 Irinotecan and vandetanib create synergies for treatment of pancreatic cancer patients with concomitant TP53 and KRAS mutations. Irinotecan 0-10 KRAS proto-oncogene, GTPase Homo sapiens 113-117 32743640-8 2021 Irinotecan and vandetanib are prospective drugs for PAAD patients with KRASG12Dmutation and TP53 mutation. Irinotecan 0-10 tumor protein p53 Homo sapiens 92-96 33930649-12 2021 Lastly, the irinotecan-induced decrease in the secretion of MMP-2 and MMP-9 by oral epithelial cells was partially restored by the green tea extract and EGCG. Irinotecan 12-22 matrix metallopeptidase 2 Homo sapiens 60-65 33930649-12 2021 Lastly, the irinotecan-induced decrease in the secretion of MMP-2 and MMP-9 by oral epithelial cells was partially restored by the green tea extract and EGCG. Irinotecan 12-22 matrix metallopeptidase 9 Homo sapiens 70-75 33970400-3 2021 OBJECTIVE: In our study, we evaluated the role of p53 expression in patients with RAS/BRAF wild-type metastatic colorectal cancer (mCRC) receiving irinotecan/cetuximab in an exploratory and a validation cohort. Irinotecan 147-157 tumor protein p53 Homo sapiens 50-53 33970400-4 2021 PATIENTS AND METHODS: p53 expression was analysed in patients with RAS/BRAF wild-type mCRC receiving second-line or third-line irinotecan/cetuximab. Irinotecan 127-137 tumor protein p53 Homo sapiens 22-25 33922657-3 2021 In this study, we demonstrated that curcumin, a plant polyphenol, sensitizes BRCA2-deficient cells to CPT-11 by impairing RAD52 recombinase in MCF7 cells. Irinotecan 102-108 BRCA2 DNA repair associated Homo sapiens 77-82 34025639-6 2021 The elevated disease activity index and histological score of colon as well as the up-regulated mRNA and protein levels of TNF-alpha, IL-1beta, and IL-6 in the colonic tissue of CPT-11-treated mice were significantly decreased by TFGU. Irinotecan 178-184 tumor necrosis factor Mus musculus 123-132 34025639-6 2021 The elevated disease activity index and histological score of colon as well as the up-regulated mRNA and protein levels of TNF-alpha, IL-1beta, and IL-6 in the colonic tissue of CPT-11-treated mice were significantly decreased by TFGU. Irinotecan 178-184 interleukin 1 alpha Mus musculus 134-142 34025639-6 2021 The elevated disease activity index and histological score of colon as well as the up-regulated mRNA and protein levels of TNF-alpha, IL-1beta, and IL-6 in the colonic tissue of CPT-11-treated mice were significantly decreased by TFGU. Irinotecan 178-184 interleukin 6 Mus musculus 148-152 34025639-10 2021 Since uric acid is a ligand of the NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome, TFGU was further validated to inhibit the activation of NLRP3 inflammasome by CPT-11. Irinotecan 186-192 NLR family, pyrin domain containing 3 Mus musculus 164-169 33677798-9 2021 Cerv54 had increased levels of CES1, which catalyzes the conversion of irinotecan to the active form, SN38, although in Cerv54 cells, SN38 was undetectable, CES1 expression and activity were markedly low compared to the liver, and a CES1 inhibitor had no effect on irinotecan sensitivity. Irinotecan 71-81 carboxylesterase 1 Homo sapiens 31-35 33677798-9 2021 Cerv54 had increased levels of CES1, which catalyzes the conversion of irinotecan to the active form, SN38, although in Cerv54 cells, SN38 was undetectable, CES1 expression and activity were markedly low compared to the liver, and a CES1 inhibitor had no effect on irinotecan sensitivity. Irinotecan 265-275 carboxylesterase 1 Homo sapiens 31-35 33907276-6 2021 We established that chemotherapy agents, particularly 7-ethyl-10-hydroxycamptothecin (SN-38), the active metabolite of irinotecan, stimulated the expression of stimulatory MHC class I alleles through stimulation the pathway of transporters associated with antigen processing 1 and 2 (TAP1 and TAP2) in cell line models. Irinotecan 54-84 transporter 1, ATP binding cassette subfamily B member Homo sapiens 284-288 33907276-6 2021 We established that chemotherapy agents, particularly 7-ethyl-10-hydroxycamptothecin (SN-38), the active metabolite of irinotecan, stimulated the expression of stimulatory MHC class I alleles through stimulation the pathway of transporters associated with antigen processing 1 and 2 (TAP1 and TAP2) in cell line models. Irinotecan 54-84 transporter 2, ATP binding cassette subfamily B member Homo sapiens 293-297 33907276-6 2021 We established that chemotherapy agents, particularly 7-ethyl-10-hydroxycamptothecin (SN-38), the active metabolite of irinotecan, stimulated the expression of stimulatory MHC class I alleles through stimulation the pathway of transporters associated with antigen processing 1 and 2 (TAP1 and TAP2) in cell line models. Irinotecan 86-91 transporter 1, ATP binding cassette subfamily B member Homo sapiens 284-288 33907276-6 2021 We established that chemotherapy agents, particularly 7-ethyl-10-hydroxycamptothecin (SN-38), the active metabolite of irinotecan, stimulated the expression of stimulatory MHC class I alleles through stimulation the pathway of transporters associated with antigen processing 1 and 2 (TAP1 and TAP2) in cell line models. Irinotecan 86-91 transporter 2, ATP binding cassette subfamily B member Homo sapiens 293-297 33907276-6 2021 We established that chemotherapy agents, particularly 7-ethyl-10-hydroxycamptothecin (SN-38), the active metabolite of irinotecan, stimulated the expression of stimulatory MHC class I alleles through stimulation the pathway of transporters associated with antigen processing 1 and 2 (TAP1 and TAP2) in cell line models. Irinotecan 119-129 transporter 1, ATP binding cassette subfamily B member Homo sapiens 284-288 33907276-6 2021 We established that chemotherapy agents, particularly 7-ethyl-10-hydroxycamptothecin (SN-38), the active metabolite of irinotecan, stimulated the expression of stimulatory MHC class I alleles through stimulation the pathway of transporters associated with antigen processing 1 and 2 (TAP1 and TAP2) in cell line models. Irinotecan 119-129 transporter 2, ATP binding cassette subfamily B member Homo sapiens 293-297 33995089-0 2021 Case Report: 7-Ethyl-10-Hydroxycamptothecin, a DNA Topoisomerase I Inhibitor, Performs BRD4 Inhibitory Activity and Inhibits Human Leukemic Cell Growth. Irinotecan 13-43 bromodomain containing 4 Homo sapiens 87-91 33995089-2 2021 In our study, SN-38 was characterized as a potent and reversible BRD4 inhibitor [IC50 = 660.2 nM against BRD4 (BD1) and IC50 = 547.7 nM against BRD4 (BD2)] in biochemical assay using drug repurposing strategy. Irinotecan 14-19 bromodomain containing 4 Homo sapiens 65-69 33995089-2 2021 In our study, SN-38 was characterized as a potent and reversible BRD4 inhibitor [IC50 = 660.2 nM against BRD4 (BD1) and IC50 = 547.7 nM against BRD4 (BD2)] in biochemical assay using drug repurposing strategy. Irinotecan 14-19 bromodomain containing 4 Homo sapiens 105-109 33995089-2 2021 In our study, SN-38 was characterized as a potent and reversible BRD4 inhibitor [IC50 = 660.2 nM against BRD4 (BD1) and IC50 = 547.7 nM against BRD4 (BD2)] in biochemical assay using drug repurposing strategy. Irinotecan 14-19 defensin beta 1 Homo sapiens 111-114 33995089-2 2021 In our study, SN-38 was characterized as a potent and reversible BRD4 inhibitor [IC50 = 660.2 nM against BRD4 (BD1) and IC50 = 547.7 nM against BRD4 (BD2)] in biochemical assay using drug repurposing strategy. Irinotecan 14-19 bromodomain containing 4 Homo sapiens 105-109 33995089-2 2021 In our study, SN-38 was characterized as a potent and reversible BRD4 inhibitor [IC50 = 660.2 nM against BRD4 (BD1) and IC50 = 547.7 nM against BRD4 (BD2)] in biochemical assay using drug repurposing strategy. Irinotecan 14-19 defensin beta 4A Homo sapiens 150-153 33995089-3 2021 Additional cellular assay suggested that SN-38 can bind BRD4 in human leukemic cell K562 and inhibit cell growth with IC50 = 0.2798 muM in a BRD4 dependent manner partially. Irinotecan 41-46 bromodomain containing 4 Homo sapiens 56-60 33995089-3 2021 Additional cellular assay suggested that SN-38 can bind BRD4 in human leukemic cell K562 and inhibit cell growth with IC50 = 0.2798 muM in a BRD4 dependent manner partially. Irinotecan 41-46 bromodomain containing 4 Homo sapiens 141-145 33995089-4 2021 Additionally, mechanism study indicated that SN-38 can induce the accumulation of BRD4 substrate c-Myc and cleavage of caspase 3. Irinotecan 45-50 bromodomain containing 4 Homo sapiens 82-86 33995089-4 2021 Additionally, mechanism study indicated that SN-38 can induce the accumulation of BRD4 substrate c-Myc and cleavage of caspase 3. Irinotecan 45-50 MYC proto-oncogene, bHLH transcription factor Homo sapiens 97-102 33995089-4 2021 Additionally, mechanism study indicated that SN-38 can induce the accumulation of BRD4 substrate c-Myc and cleavage of caspase 3. Irinotecan 45-50 caspase 3 Homo sapiens 119-128 33995089-5 2021 In sum, our findings identified BRD4 as a new target of SN-38 and reveals SN-38 as a modifier of histone acetylation reader for the first time, which may provide a new insight for further optimization of dual target inhibitor. Irinotecan 56-61 bromodomain containing 4 Homo sapiens 32-36 33888518-12 2021 Conclusion AREG/EREG IHC identified patients who benefitted from the addition of panitumumab to irinotecan chemotherapy. Irinotecan 96-106 amphiregulin Homo sapiens 11-15 33888518-12 2021 Conclusion AREG/EREG IHC identified patients who benefitted from the addition of panitumumab to irinotecan chemotherapy. Irinotecan 96-106 epiregulin Homo sapiens 16-20 33922657-6 2021 In vivo, xenograft model studies showed that curcumin combined with CPT-11 reduced the growth of BRCA2-knockout MCF7 tumors but not MCF7 tumors. Irinotecan 68-74 BRCA2 DNA repair associated Homo sapiens 97-102 33837924-7 2021 Reports for UGT1A1-metabolized irinotecan and the CYP2C19-metabolized drugs voriconazole, escitalopram and clopidogrel were selected for comparison between the subgroups based upon known genetic polymorphisms with high prevalence in Japan. Irinotecan 31-41 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-18 33882206-2 2021 Sacituzumab govitecan is an antibody-drug conjugate composed of an antibody targeting the human trophoblast cell-surface antigen 2 (Trop-2), which is expressed in the majority of breast cancers, coupled to SN-38 (topoisomerase I inhibitor) through a proprietary hydrolyzable linker. Irinotecan 206-211 tumor associated calcium signal transducer 2 Homo sapiens 132-138 32981006-1 2021 Camptothecin (CPT) and its derivatives, irinotecan and topotecan are specific topoisomerase I (Top1) inhibitors and potent anticancer drugs. Irinotecan 40-50 DNA topoisomerase I Homo sapiens 95-99 33872865-6 2021 Pasta containing 20% lupin flour and 10% RS4 had higher gelatinization onset temperature values, and lower enthalpy than 100% semolina pasta. Irinotecan 41-44 solute carrier family 45 member 1 Homo sapiens 0-5 33795652-7 2021 Lastly, we preliminary explored the translational effect of HOXC6 and found that silencing of HOXC6 made HCT116 and HT29 cells more sensitive to irinotecan. Irinotecan 145-155 homeobox C6 Homo sapiens 94-99 33475937-7 2021 Further results revealed that the downregulation of the efflux transporter BCRP by long-term fractionated irradiation was an important factor contributing to the increased cytotoxicity of SN-38. Irinotecan 188-193 BCR pseudogene 1 Homo sapiens 75-79 33462346-2 2021 DPYD and UGT1A1 variants are relevant predictors of fluoropyrimidine and irinotecan-associated adverse events (AEs). Irinotecan 73-83 dihydropyrimidine dehydrogenase Homo sapiens 0-4 33593942-1 2021 CF10 is a 2nd generation polymeric fluoropyrimidine (FP) that targets both thymidylate synthase (TS), the target of 5-fluorouracil (5-FU), and DNA topoisomerase 1 (Top1), the target of irinotecan, two drugs that are key components of FOLFIRNOX, a standard of care regimen for pancreatic ductal adenocarcinoma (PDAC). Irinotecan 185-195 thymidylate synthetase Homo sapiens 97-99 33593942-1 2021 CF10 is a 2nd generation polymeric fluoropyrimidine (FP) that targets both thymidylate synthase (TS), the target of 5-fluorouracil (5-FU), and DNA topoisomerase 1 (Top1), the target of irinotecan, two drugs that are key components of FOLFIRNOX, a standard of care regimen for pancreatic ductal adenocarcinoma (PDAC). Irinotecan 185-195 DNA topoisomerase I Homo sapiens 164-168 33733372-3 2021 METHODS: Two pharmacokinetic and one myelosuppression model were assembled to predict concentrations of irinotecan and its metabolite SN-38 given different UGT1A1 genotypes (poor metabolizers: CLSN-38: -36%) and neutropenia following conventional versus PGx-based dosing (350 versus 245 mg/m2 (-30%)). Irinotecan 104-114 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 156-162 33462346-2 2021 DPYD and UGT1A1 variants are relevant predictors of fluoropyrimidine and irinotecan-associated adverse events (AEs). Irinotecan 73-83 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 9-15 33733372-3 2021 METHODS: Two pharmacokinetic and one myelosuppression model were assembled to predict concentrations of irinotecan and its metabolite SN-38 given different UGT1A1 genotypes (poor metabolizers: CLSN-38: -36%) and neutropenia following conventional versus PGx-based dosing (350 versus 245 mg/m2 (-30%)). Irinotecan 134-139 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 156-162 33462346-9 2021 DPYD IVS14+1A allele is confirmed to be associated with fluoropyrimidine life-threatening toxicities, and UGT1A1*28 is related with a higher risk of hematologic AEs following irinotecan treatment. Irinotecan 175-185 dihydropyrimidine dehydrogenase Homo sapiens 0-4 33462346-9 2021 DPYD IVS14+1A allele is confirmed to be associated with fluoropyrimidine life-threatening toxicities, and UGT1A1*28 is related with a higher risk of hematologic AEs following irinotecan treatment. Irinotecan 175-185 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 106-112 33869031-10 2021 In PDX models with mutated p53 status, there was significant tumor growth inhibition from the combination of AZD1775 with irinotecan or capecitabine (P <= 0.03), while PDX models with wild type p53 did not show anti-tumor synergy from the same combinations (P >= 0.08). Irinotecan 122-132 tumor protein p53 Homo sapiens 27-30 33790625-0 2021 Influence of UGT1A1 *6/*28 Polymorphisms on Irinotecan-Related Toxicity and Survival in Pediatric Patients with Relapsed/Refractory Solid Tumors Treated with the VIT Regimen. Irinotecan 44-54 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 13-19 33790625-0 2021 Influence of UGT1A1 *6/*28 Polymorphisms on Irinotecan-Related Toxicity and Survival in Pediatric Patients with Relapsed/Refractory Solid Tumors Treated with the VIT Regimen. Irinotecan 44-54 vitrin Homo sapiens 162-165 33790625-1 2021 Objective: The association between UGT1A1*6/*28 polymorphisms and treatment outcomes of irinotecan in children remains unknown. Irinotecan 88-98 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 35-41 33790625-2 2021 This retrospective study investigated the influence of UGT1A1*6/*28 polymorphisms on irinotecan toxicity and survival of pediatric patients with relapsed/refractory solid tumors. Irinotecan 85-95 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 55-61 33790625-8 2021 Moreover, patients with mutant UGT1A1*6 genotypes were more likely to develop grade I-IV irinotecan-related diarrhea (P = 0.043) and anemia (P = 0.002). Irinotecan 89-99 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 31-37 33790625-9 2021 Overall, the UGT1A1*28 polymorphism may play a protective role against irinotecan-related diarrhea and abdominal pain. Irinotecan 71-81 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 13-19 33790625-10 2021 Conclusion: In relapsed/refractory pediatric solid tumors, the UGT1A1*6 polymorphism was a risk factor of irinotecan-related diarrhea and anemia. Irinotecan 106-116 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 63-69 33790625-11 2021 The UGT1A1*28 polymorphism may serve a protective role in irinotecan-related abdominal pain and diarrhea. Irinotecan 58-68 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 33664385-0 2021 Entropy-driven binding of gut bacterial beta-glucuronidase inhibitors ameliorates irinotecan-induced toxicity. Irinotecan 82-92 glucuronidase beta Homo sapiens 40-58 33630990-1 2021 Carboxylesterase 2 (CES 2) is a key enzyme in the activation of the prodrug irinotecan (CPT-11) in the treatment against colorectal cancer and also has some relationship with the side effect of CPT-11 in clinical applications. Irinotecan 88-94 carboxylesterase 2 Homo sapiens 20-25 33630990-1 2021 Carboxylesterase 2 (CES 2) is a key enzyme in the activation of the prodrug irinotecan (CPT-11) in the treatment against colorectal cancer and also has some relationship with the side effect of CPT-11 in clinical applications. Irinotecan 194-200 carboxylesterase 2 Homo sapiens 0-18 33630990-1 2021 Carboxylesterase 2 (CES 2) is a key enzyme in the activation of the prodrug irinotecan (CPT-11) in the treatment against colorectal cancer and also has some relationship with the side effect of CPT-11 in clinical applications. Irinotecan 194-200 carboxylesterase 2 Homo sapiens 20-25 33728696-0 2021 UGT1A1 Gene Polymorphisms in Patients with Small Cell Lung Cancer Treated with Irinotecan-Platinum Doublet Chemotherapy and Their Association with Gastrointestinal Toxicity and Overall Survival. Irinotecan 79-89 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 33728696-1 2021 BACKGROUND: Irinotecan (CPT11) is an important drug for small cell lung cancer (SCLC) chemotherapy (CTx). Irinotecan 12-22 cytochrome P450 family 27 subfamily A member 1 Homo sapiens 100-103 33728696-1 2021 BACKGROUND: Irinotecan (CPT11) is an important drug for small cell lung cancer (SCLC) chemotherapy (CTx). Irinotecan 24-29 cytochrome P450 family 27 subfamily A member 1 Homo sapiens 100-103 33728696-18 2021 IMPLICATIONS FOR PRACTICE: UGT1A1 gene polymorphisms are known to influence irinotecan-related toxicity. Irinotecan 76-86 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 27-33 33728696-19 2021 In this prospective cohort study of patients with small cell lung cancer receiving first-line irinotecan-platinum chemotherapy, the prevalence of UGT1A1*6 polymorphisms was found to be 10.8% UGT1A1*6 and 58.8% UGT1A1*28 homo/heterozygous mutant, respectively. Irinotecan 94-104 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 146-152 33728696-19 2021 In this prospective cohort study of patients with small cell lung cancer receiving first-line irinotecan-platinum chemotherapy, the prevalence of UGT1A1*6 polymorphisms was found to be 10.8% UGT1A1*6 and 58.8% UGT1A1*28 homo/heterozygous mutant, respectively. Irinotecan 94-104 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 191-197 33728696-19 2021 In this prospective cohort study of patients with small cell lung cancer receiving first-line irinotecan-platinum chemotherapy, the prevalence of UGT1A1*6 polymorphisms was found to be 10.8% UGT1A1*6 and 58.8% UGT1A1*28 homo/heterozygous mutant, respectively. Irinotecan 94-104 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 191-197 33630990-1 2021 Carboxylesterase 2 (CES 2) is a key enzyme in the activation of the prodrug irinotecan (CPT-11) in the treatment against colorectal cancer and also has some relationship with the side effect of CPT-11 in clinical applications. Irinotecan 76-86 carboxylesterase 2 Homo sapiens 0-18 33630990-1 2021 Carboxylesterase 2 (CES 2) is a key enzyme in the activation of the prodrug irinotecan (CPT-11) in the treatment against colorectal cancer and also has some relationship with the side effect of CPT-11 in clinical applications. Irinotecan 76-86 carboxylesterase 2 Homo sapiens 20-25 33630990-1 2021 Carboxylesterase 2 (CES 2) is a key enzyme in the activation of the prodrug irinotecan (CPT-11) in the treatment against colorectal cancer and also has some relationship with the side effect of CPT-11 in clinical applications. Irinotecan 88-94 carboxylesterase 2 Homo sapiens 0-18 33662636-10 2021 In addition, ZFX expression was negatively correlated with the sensitivity of fresh ESCC tissues to chemotherapeutic drugs, including cisplatin, docetaxel, fluorouracil, and irinotecan. Irinotecan 174-184 zinc finger protein X-linked Homo sapiens 13-16 33569738-1 2021 INTRODUCTION: Recently the phase 3 BEACON trial showed that the combination of encorafenib, cetuximab, and binimetinib versus cetuximab and irinotecan/FOLFIRI improved overall survival in pre-treated patients with metastatic colorectal cancer (mCRC) with BRAF V600E mutation. Irinotecan 140-150 ubiquitin like 5 Homo sapiens 35-41 33386925-9 2021 CONCLUSION: Based on our findings, we recommend that in Japanese advanced PC patients with UGT1A1- DV treated with FOLFIRINOX, irinotecan be administered at an initial dose of <= 120 mg/m2. Irinotecan 127-137 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 91-97 33323683-5 2021 MGMT expression also correlated significantly with resistance to TMZ+irinotecan (IRN) (in-vitro as the SN38 active metabolite). Irinotecan 69-79 O-6-methylguanine-DNA methyltransferase Homo sapiens 0-4 33386925-1 2021 BACKGROUND: UGT1A1 *28 and *6 polymorphism is associated with reduced enzyme activity and severe toxicities of irinotecan, especially in patients with homozygous or heterozygous for UGT1A1*28 or *6 polymorphism for both UGT1A1*28 and *6 (double-variant-type of UGT1A1 polymorphism, UGT1A1-DV). Irinotecan 111-121 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-18 33386925-1 2021 BACKGROUND: UGT1A1 *28 and *6 polymorphism is associated with reduced enzyme activity and severe toxicities of irinotecan, especially in patients with homozygous or heterozygous for UGT1A1*28 or *6 polymorphism for both UGT1A1*28 and *6 (double-variant-type of UGT1A1 polymorphism, UGT1A1-DV). Irinotecan 111-121 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 182-188 33386925-1 2021 BACKGROUND: UGT1A1 *28 and *6 polymorphism is associated with reduced enzyme activity and severe toxicities of irinotecan, especially in patients with homozygous or heterozygous for UGT1A1*28 or *6 polymorphism for both UGT1A1*28 and *6 (double-variant-type of UGT1A1 polymorphism, UGT1A1-DV). Irinotecan 111-121 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 182-188 33386925-1 2021 BACKGROUND: UGT1A1 *28 and *6 polymorphism is associated with reduced enzyme activity and severe toxicities of irinotecan, especially in patients with homozygous or heterozygous for UGT1A1*28 or *6 polymorphism for both UGT1A1*28 and *6 (double-variant-type of UGT1A1 polymorphism, UGT1A1-DV). Irinotecan 111-121 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 182-188 33386925-1 2021 BACKGROUND: UGT1A1 *28 and *6 polymorphism is associated with reduced enzyme activity and severe toxicities of irinotecan, especially in patients with homozygous or heterozygous for UGT1A1*28 or *6 polymorphism for both UGT1A1*28 and *6 (double-variant-type of UGT1A1 polymorphism, UGT1A1-DV). Irinotecan 111-121 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 182-188 33386925-3 2021 The optimal dose of irinotecan as a component of the FOLFIRINOX has not been established yet for patients with UGT1A1-DV. Irinotecan 20-30 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 111-117 33386925-5 2021 We evaluated the patient characteristics, efficacy and safety of FOLFIRINOX and investigate the optimal initial dose of irinotecan in Japanese advanced PC patients with UGT1A1-DV. Irinotecan 120-130 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 169-175 33531790-10 2021 The bioinformatic analysis revealed a higher binding efficiency of CPT and its derivatives, toptecan and irinotecan against Mpro and RdRp. Irinotecan 105-115 NEWENTRY Severe acute respiratory syndrome-related coronavirus 124-128 33619631-0 2021 Effect of UGT1A1, CYP3A and CES Activities on the Pharmacokinetics of Irinotecan and its Metabolites in Patients with UGT1A1 Gene Polymorphisms. Irinotecan 70-80 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 10-16 33619631-0 2021 Effect of UGT1A1, CYP3A and CES Activities on the Pharmacokinetics of Irinotecan and its Metabolites in Patients with UGT1A1 Gene Polymorphisms. Irinotecan 70-80 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 18-23 33619631-0 2021 Effect of UGT1A1, CYP3A and CES Activities on the Pharmacokinetics of Irinotecan and its Metabolites in Patients with UGT1A1 Gene Polymorphisms. Irinotecan 70-80 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 118-124 33619631-2 2021 SN-38 is then converted to the inactive metabolite SN-38 glucuronide (SN-38G) by glucuronosyltransferase 1A1 (UGT1A1). Irinotecan 0-5 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 110-116 33619631-3 2021 Genetic polymorphisms in UGT1A1 have been associated with altered SN-38 pharmacokinetics, which increase the risk of toxicity in patients. Irinotecan 66-71 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 25-31 33619631-4 2021 CPT-11 is also converted to 7-ethyl-10-[4-N-(5-aminopentanoic acid)-1-piperidino]carbonyloxycamptothecin (APC) and 7-ethyl-10-(4-amino-1-piperidino) carbonyloxycamptothecin (NPC) by cytochrome P450 3A (CYP3A), and this route also affects the plasma concentration of SN-38. Irinotecan 0-6 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 182-200 33619631-4 2021 CPT-11 is also converted to 7-ethyl-10-[4-N-(5-aminopentanoic acid)-1-piperidino]carbonyloxycamptothecin (APC) and 7-ethyl-10-(4-amino-1-piperidino) carbonyloxycamptothecin (NPC) by cytochrome P450 3A (CYP3A), and this route also affects the plasma concentration of SN-38. Irinotecan 0-6 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 202-207 33619631-5 2021 We evaluated the activities of UGT1A1, CYP3A, and CES and the factors affecting the pharmacokinetics of plasma SN-38 in patients with UGT1A1 gene polymorphisms. Irinotecan 111-116 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 134-140 33619631-12 2021 Since UGT1A1 activity was low in patient 1 with a high AUC of SN-38, it can be said that the increase in plasma concentration was due to a decrease in UGT1A1 activity. Irinotecan 62-67 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 6-12 33619631-14 2021 Patient 3 had the lowest AUC of SN-38, caused by a lower level of CES activity and increased CYP3A activity. Irinotecan 32-37 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 93-98 33619631-15 2021 CONCLUSION: In this study, we indicated that the plasma AUC of SN-38 and AUC ratio of SN-38G/SN-38 may depend on changes in the activities of CYP3A, CES, and UGT1A1. Irinotecan 63-68 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 142-147 33619631-15 2021 CONCLUSION: In this study, we indicated that the plasma AUC of SN-38 and AUC ratio of SN-38G/SN-38 may depend on changes in the activities of CYP3A, CES, and UGT1A1. Irinotecan 63-68 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 158-164 33619631-15 2021 CONCLUSION: In this study, we indicated that the plasma AUC of SN-38 and AUC ratio of SN-38G/SN-38 may depend on changes in the activities of CYP3A, CES, and UGT1A1. Irinotecan 86-91 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 142-147 33619631-15 2021 CONCLUSION: In this study, we indicated that the plasma AUC of SN-38 and AUC ratio of SN-38G/SN-38 may depend on changes in the activities of CYP3A, CES, and UGT1A1. Irinotecan 86-91 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 158-164 33531790-10 2021 The bioinformatic analysis revealed a higher binding efficiency of CPT and its derivatives, toptecan and irinotecan against Mpro and RdRp. Irinotecan 105-115 ORF1a polyprotein;ORF1ab polyprotein Severe acute respiratory syndrome coronavirus 2 133-137 33912512-1 2021 Background: This study investigated the inhibition of tumor growth in castrate-resistant prostate cancer (CRPC)-bearing mice by tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-overexpressing adipose-derived stem cells (ADSCs) (hTERT-ADSC.sTRAIL), which was enhanced by combined treatment with CPT-11. Irinotecan 309-315 tumor necrosis factor (ligand) superfamily, member 10 Mus musculus 128-183 33574948-7 2021 AKT1/CTNNB1 mutations were also associated with oxaliplatin, irinotecan, SN-38 and 5-fluorouracil resistance. Irinotecan 61-71 AKT serine/threonine kinase 1 Homo sapiens 0-4 33574948-7 2021 AKT1/CTNNB1 mutations were also associated with oxaliplatin, irinotecan, SN-38 and 5-fluorouracil resistance. Irinotecan 61-71 catenin beta 1 Homo sapiens 5-11 33574948-7 2021 AKT1/CTNNB1 mutations were also associated with oxaliplatin, irinotecan, SN-38 and 5-fluorouracil resistance. Irinotecan 73-78 AKT serine/threonine kinase 1 Homo sapiens 0-4 33574948-7 2021 AKT1/CTNNB1 mutations were also associated with oxaliplatin, irinotecan, SN-38 and 5-fluorouracil resistance. Irinotecan 73-78 catenin beta 1 Homo sapiens 5-11 33912512-1 2021 Background: This study investigated the inhibition of tumor growth in castrate-resistant prostate cancer (CRPC)-bearing mice by tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-overexpressing adipose-derived stem cells (ADSCs) (hTERT-ADSC.sTRAIL), which was enhanced by combined treatment with CPT-11. Irinotecan 309-315 tumor necrosis factor (ligand) superfamily, member 10 Mus musculus 185-190 33912512-12 2021 In the in vivo study, the inhibition of tumor growth in CRPC-bearing mice by TRAIL-overexpressing adipose stem cells was enhanced by combined treatment with the chemotherapeutic agent CPT-11 compared with that in the treatment with cpt-11 alone. Irinotecan 184-190 tumor necrosis factor (ligand) superfamily, member 10 Mus musculus 77-82 33912512-12 2021 In the in vivo study, the inhibition of tumor growth in CRPC-bearing mice by TRAIL-overexpressing adipose stem cells was enhanced by combined treatment with the chemotherapeutic agent CPT-11 compared with that in the treatment with cpt-11 alone. Irinotecan 232-238 tumor necrosis factor (ligand) superfamily, member 10 Mus musculus 77-82 33291124-1 2021 Human carboxylesterase 2 (hCES2A) is a key target to ameliorate the intestinal toxicity triggered by irinotecan that causes severe diarrhea in 50%-80% of patients receiving this anticancer agent. Irinotecan 101-111 carboxylesterase 2 Homo sapiens 6-24 33568030-11 2021 CONCLUSION: Clotrimazole displayed a strong inhibitory effect against hCE2, which might be used as a potential combined agent co-administrated with CPT-11 to alleviate the hCE2-mediated severe side effects. Irinotecan 148-154 carboxylesterase 2 Homo sapiens 70-74 33568030-11 2021 CONCLUSION: Clotrimazole displayed a strong inhibitory effect against hCE2, which might be used as a potential combined agent co-administrated with CPT-11 to alleviate the hCE2-mediated severe side effects. Irinotecan 148-154 carboxylesterase 2 Homo sapiens 172-176 33089597-2 2021 Here, we determined whether an inhibitor of EZH2 enhanced the effect of standard of care chemotherapeutic agents: irinotecan, vincristine, and cyclophosphamide. Irinotecan 114-124 enhancer of zeste 2 polycomb repressive complex 2 subunit Homo sapiens 44-48 33615175-9 2021 Further, formation of glucuronides of raloxifene and 7-ethyl-10-hydroxycamptothecin were reduced in the cells expressing the UGT1A1 P152T mutant. Irinotecan 53-83 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 125-131 33526988-5 2021 Thus, this study was aimed to examine whether MutS homolog (MSH) 2, one member of DNA repair system, plays a role to affect the cytotoxicity of CPT-11 to CRCs. Irinotecan 144-150 mutS homolog 2 Homo sapiens 46-66 33526988-7 2021 It was shown that MSH2 gene and protein expression could be upregulated in DLD-1 cells under CPT-11 treatment and this upregulation subsequently attenuates the sensitivity of DLD-1 cells to CPT-11. Irinotecan 93-99 mutS homolog 2 Homo sapiens 18-22 33526988-7 2021 It was shown that MSH2 gene and protein expression could be upregulated in DLD-1 cells under CPT-11 treatment and this upregulation subsequently attenuates the sensitivity of DLD-1 cells to CPT-11. Irinotecan 190-196 mutS homolog 2 Homo sapiens 18-22 33526988-9 2021 These results elucidate the drug resistance role of MSH2 upregulation in the CPT-11-treated DLD-1 CRC cells. Irinotecan 77-83 mutS homolog 2 Homo sapiens 52-56 33692943-4 2020 In the present study, we established a novel irinotecan-resistant human colon cell line to investigate the underlying mechanism(s) of irinotecan resistance, particularly the overexpression of ABC transporters. Irinotecan 45-55 ATP binding cassette subfamily B member 6 (Langereis blood group) Homo sapiens 192-195 33692943-4 2020 In the present study, we established a novel irinotecan-resistant human colon cell line to investigate the underlying mechanism(s) of irinotecan resistance, particularly the overexpression of ABC transporters. Irinotecan 134-144 ATP binding cassette subfamily B member 6 (Langereis blood group) Homo sapiens 192-195 33692943-10 2020 This finding was further confirmed by reversal studies that inhibiting efflux function of ABCG2 was able to completely abolish drug resistance to irinotecan as well as other ABCG2 substrates in S1-IR20 cells. Irinotecan 146-156 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 90-95 33692943-11 2020 In conclusion, our work established an in vitro model of irinotecan resistance in CRC and suggested ABCG2 overexpression as one of the underlying mechanisms of acquired resistance to irinotecan. Irinotecan 183-193 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 100-105 33611805-1 2021 BACKGROUND: Both protracted irinotecan and antiangiogenesis therapy have shown promising efficacy against Ewing sarcoma (EWS). Irinotecan 28-38 EWS RNA binding protein 1 Homo sapiens 121-124 33611805-15 2021 CONCLUSION: The combination of vincristine, irinotecan, and anlotinib demonstrated an acceptable toxicity profile and promising clinical efficacy in patients with advanced EWS. Irinotecan 44-54 EWS RNA binding protein 1 Homo sapiens 172-175 33611805-16 2021 IMPLICATIONS FOR PRACTICE: This is the first trial to evaluate an irinotecan-based regimen in combination with antiangiogenesis tyrosine kinase inhibitors in Ewing sarcoma (EWS). Irinotecan 66-76 EWS RNA binding protein 1 Homo sapiens 173-176 33611805-21 2021 This study shows that the combination of vincristine, irinotecan, and anlotinib demonstrates an acceptable toxicity profile and promising clinical efficacy in patients with advanced EWS. Irinotecan 54-64 EWS RNA binding protein 1 Homo sapiens 182-185 33568030-9 2021 Furthermore, the inhibitions of clotrimazole towards hCE2-mediated hydrolysis of FD, NCEN and CPT-11 were all in competitive mode with the Ki values of 0.483 muM, 8.63 muM and 29.0 muM, respectively. Irinotecan 94-100 carboxylesterase 2 Homo sapiens 53-57 32840656-0 2021 The introduction of immunosuppressor (TDO inhibitor) significantly improved the efficacy of irinotecan in treating hepatocellular carcinoma. Irinotecan 92-102 tryptophan 2,3-dioxygenase Homo sapiens 38-41 32840656-1 2021 As TDO inhibitors can improve the efficacy of tumor chemotherapeutics, two TDO-targeted conjugates consisting of irinotecan (Ir) and a TDO inhibitor unit were designed and prepared to reverse tumor immune suppression, which could remarkably enhance antitumor activity of Ir by boosting cellular uptakes against TDO overexpressed HepG2 cancer cells. Irinotecan 113-123 tryptophan 2,3-dioxygenase Homo sapiens 3-6 32840656-1 2021 As TDO inhibitors can improve the efficacy of tumor chemotherapeutics, two TDO-targeted conjugates consisting of irinotecan (Ir) and a TDO inhibitor unit were designed and prepared to reverse tumor immune suppression, which could remarkably enhance antitumor activity of Ir by boosting cellular uptakes against TDO overexpressed HepG2 cancer cells. Irinotecan 113-123 tryptophan 2,3-dioxygenase Homo sapiens 75-78 32840656-1 2021 As TDO inhibitors can improve the efficacy of tumor chemotherapeutics, two TDO-targeted conjugates consisting of irinotecan (Ir) and a TDO inhibitor unit were designed and prepared to reverse tumor immune suppression, which could remarkably enhance antitumor activity of Ir by boosting cellular uptakes against TDO overexpressed HepG2 cancer cells. Irinotecan 113-123 tryptophan 2,3-dioxygenase Homo sapiens 75-78 32840656-1 2021 As TDO inhibitors can improve the efficacy of tumor chemotherapeutics, two TDO-targeted conjugates consisting of irinotecan (Ir) and a TDO inhibitor unit were designed and prepared to reverse tumor immune suppression, which could remarkably enhance antitumor activity of Ir by boosting cellular uptakes against TDO overexpressed HepG2 cancer cells. Irinotecan 113-123 tryptophan 2,3-dioxygenase Homo sapiens 75-78 32840656-7 2021 Conjugates obtained by combining an immune checkpoint TDO inhibitor with irinotecan via different linkers could improve tumor immune microenvironment by inhibiting the TDO enzyme expression to block kynurenine production and induce HepG2 cancer cell apoptosis via DNA damage through releasing a TDO inhibitor and irinotecan in cancer cells. Irinotecan 73-83 tryptophan 2,3-dioxygenase Homo sapiens 168-171 32840656-7 2021 Conjugates obtained by combining an immune checkpoint TDO inhibitor with irinotecan via different linkers could improve tumor immune microenvironment by inhibiting the TDO enzyme expression to block kynurenine production and induce HepG2 cancer cell apoptosis via DNA damage through releasing a TDO inhibitor and irinotecan in cancer cells. Irinotecan 73-83 tryptophan 2,3-dioxygenase Homo sapiens 168-171 32840656-7 2021 Conjugates obtained by combining an immune checkpoint TDO inhibitor with irinotecan via different linkers could improve tumor immune microenvironment by inhibiting the TDO enzyme expression to block kynurenine production and induce HepG2 cancer cell apoptosis via DNA damage through releasing a TDO inhibitor and irinotecan in cancer cells. Irinotecan 313-323 tryptophan 2,3-dioxygenase Homo sapiens 54-57 32840656-7 2021 Conjugates obtained by combining an immune checkpoint TDO inhibitor with irinotecan via different linkers could improve tumor immune microenvironment by inhibiting the TDO enzyme expression to block kynurenine production and induce HepG2 cancer cell apoptosis via DNA damage through releasing a TDO inhibitor and irinotecan in cancer cells. Irinotecan 313-323 tryptophan 2,3-dioxygenase Homo sapiens 168-171 32840656-7 2021 Conjugates obtained by combining an immune checkpoint TDO inhibitor with irinotecan via different linkers could improve tumor immune microenvironment by inhibiting the TDO enzyme expression to block kynurenine production and induce HepG2 cancer cell apoptosis via DNA damage through releasing a TDO inhibitor and irinotecan in cancer cells. Irinotecan 313-323 tryptophan 2,3-dioxygenase Homo sapiens 168-171 33039558-6 2021 STAG3 knockdown inhibited cell migration and increased drug sensitivity to oxaliplatin, 5-fluorouracil, irinotecan hydrochloride hydrate, and BRAF inhibitor in CRC cell lines. Irinotecan 104-136 stromal antigen 3 Homo sapiens 0-5 33747719-2 2021 It is demonstrated that improved irinotecan delivery by a lipid bilayer coated mesoporous silica nanoparticle, also known as a silicasome, can improve PDAC survival through a chemo-immunotherapy response in an orthotopic Kras-dependent pancreatic cancer model. Irinotecan 33-43 KRAS proto-oncogene, GTPase Homo sapiens 221-225 33747719-6 2021 The improved delivery of irinotecan by the silicasome is accompanied by robust antitumor immunity, which can be synergistically enhanced by anti-PD-1 in the orthotopic model. Irinotecan 25-35 programmed cell death 1 Homo sapiens 145-149 33367449-6 2021 We fully validated the assay for quality control of the irinotecan solution in the injection dosage form: the standard curve was linear over the concentration range of 0.90 to at least 37.00 mug ml-1. Irinotecan 56-66 interleukin 17F Homo sapiens 195-199 33500629-0 2021 Gastric Cancer Harboring an ERBB3 Mutation Treated with a Pyrotinib-Irinotecan Combo: A Case Study. Irinotecan 68-78 erb-b2 receptor tyrosine kinase 3 Homo sapiens 28-33 33500629-6 2021 Therefore, the combined administration of irinotecan and pyrotinib may improve the clinical condition of patients with gastric cancer harboring an ERBB3 mutation. Irinotecan 42-52 erb-b2 receptor tyrosine kinase 3 Homo sapiens 147-152 33407715-0 2021 Vulnerability to low-dose combination of irinotecan and niraparib in ATM-mutated colorectal cancer. Irinotecan 41-51 ATM serine/threonine kinase Homo sapiens 69-72 33407715-12 2021 CONCLUSIONS: This work demonstrates that a numerically relevant subset of CRCs carrying heterozygous ATM mutations may benefit from the combination treatment with low doses of niraparib and irinotecan, suggesting a new potential approach in the treatment of ATM-mutated CRC, that deserves to be prospectively validated in clinical trials. Irinotecan 190-200 ATM serine/threonine kinase Homo sapiens 101-104 33407715-12 2021 CONCLUSIONS: This work demonstrates that a numerically relevant subset of CRCs carrying heterozygous ATM mutations may benefit from the combination treatment with low doses of niraparib and irinotecan, suggesting a new potential approach in the treatment of ATM-mutated CRC, that deserves to be prospectively validated in clinical trials. Irinotecan 190-200 ATM serine/threonine kinase Homo sapiens 258-261 33249316-4 2021 Through in vitro screening among several DNA-targeting chemo agents, we identified 7-ethyl-10-hydroxycamptothecin (SN38) as the most potent drug for stimulating interferon (IFN)-beta secretion and proved that this process is mediated by the passage of DNA-containing exosomes from treated tumor cells to bone marrow-derived dendritic cells (BMDCs) and subsequent activation of the STING pathway. Irinotecan 83-113 IFN1@ Homo sapiens 161-182 33569416-3 2021 Adenosine triphosphate (ATP)-binding cassette sub-family G member 2 (ABCG2) has been found highly expressed in CRC patients with poor responsiveness to folinic acid/5-FU/irinotecan. Irinotecan 170-180 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 0-67 33569416-3 2021 Adenosine triphosphate (ATP)-binding cassette sub-family G member 2 (ABCG2) has been found highly expressed in CRC patients with poor responsiveness to folinic acid/5-FU/irinotecan. Irinotecan 170-180 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 69-74 33249316-4 2021 Through in vitro screening among several DNA-targeting chemo agents, we identified 7-ethyl-10-hydroxycamptothecin (SN38) as the most potent drug for stimulating interferon (IFN)-beta secretion and proved that this process is mediated by the passage of DNA-containing exosomes from treated tumor cells to bone marrow-derived dendritic cells (BMDCs) and subsequent activation of the STING pathway. Irinotecan 115-119 IFN1@ Homo sapiens 161-182 33390536-9 2021 BCRP mRNA and protein levels were both decreased in Snail-expressing cells, which showed an increase in the intracellular accumulation of 7-ethyl-10-hydroxycamptothecin (SN-38), a BCRP substrate, resulting in reduced viability. Irinotecan 138-168 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 0-4 33468749-4 2021 NAC with FOLFOXIRI(5-fluorouracil/oxaliplatin/leucovorin/irinotecan)plus bevacizumab(BEV)was inisiated as NAC. Irinotecan 57-67 synuclein alpha Homo sapiens 0-3 33390536-9 2021 BCRP mRNA and protein levels were both decreased in Snail-expressing cells, which showed an increase in the intracellular accumulation of 7-ethyl-10-hydroxycamptothecin (SN-38), a BCRP substrate, resulting in reduced viability. Irinotecan 138-168 snail family transcriptional repressor 1 Homo sapiens 52-57 33390536-9 2021 BCRP mRNA and protein levels were both decreased in Snail-expressing cells, which showed an increase in the intracellular accumulation of 7-ethyl-10-hydroxycamptothecin (SN-38), a BCRP substrate, resulting in reduced viability. Irinotecan 138-168 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 180-184 33390536-9 2021 BCRP mRNA and protein levels were both decreased in Snail-expressing cells, which showed an increase in the intracellular accumulation of 7-ethyl-10-hydroxycamptothecin (SN-38), a BCRP substrate, resulting in reduced viability. Irinotecan 170-175 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 0-4 33390536-9 2021 BCRP mRNA and protein levels were both decreased in Snail-expressing cells, which showed an increase in the intracellular accumulation of 7-ethyl-10-hydroxycamptothecin (SN-38), a BCRP substrate, resulting in reduced viability. Irinotecan 170-175 snail family transcriptional repressor 1 Homo sapiens 52-57 33390536-9 2021 BCRP mRNA and protein levels were both decreased in Snail-expressing cells, which showed an increase in the intracellular accumulation of 7-ethyl-10-hydroxycamptothecin (SN-38), a BCRP substrate, resulting in reduced viability. Irinotecan 170-175 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 180-184 33402591-4 2021 Uridine diphosphate glucuronosyltransferase (UGT) gene polymorphisms such as UGT1A1*28/*6, age, performance status, serum lactate dehydrogenase levels, and bilirubin levels could be important considerations for predicting outcomes and tolerability with irinotecan-based regimens. Irinotecan 253-263 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 0-43 33377131-6 2020 Susceptible tumors are characterized by a stemness signature, an activated interferon pathway, and suppression of PD-1 ligands in response to 5-FU+irinotecan. Irinotecan 147-157 programmed cell death 1 Homo sapiens 114-118 33490922-5 2021 Topoisomerase 1 (TOP1) inhibitors such as irinotecan enhance cisplatin-induced SASP, which depends on the TOP1 cleavage complex-regulated cGAS pathway. Irinotecan 42-52 cyclic GMP-AMP synthase Mus musculus 138-142 33119477-1 2020 PURPOSE: Differentiating the irinotecan dose on the basis of the uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) genotype improves the pathologic complete response (pCR) rate. Irinotecan 29-39 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 65-112 33119477-1 2020 PURPOSE: Differentiating the irinotecan dose on the basis of the uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) genotype improves the pathologic complete response (pCR) rate. Irinotecan 29-39 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 114-120 33119477-14 2020 CONCLUSION: Adding irinotecan guided by UGT1A1 genotype to capecitabine-based neoadjuvant chemoradiotherapy significantly increased complete tumor response in Chinese patients. Irinotecan 19-29 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 40-46 32930615-6 2020 For example, graphene oxide incorporated with PEG and loaded with SN 38 for camptothecin analolgue as anticancer drug and revealed high cytotoxicity has an effect of 1000 times better effect than CPT in HCT-116 cells. Irinotecan 66-71 choline phosphotransferase 1 Homo sapiens 196-199 33348673-0 2020 The Paradoxical Effect of PARP Inhibitor BGP-15 on Irinotecan-Induced Cachexia and Skeletal Muscle Dysfunction. Irinotecan 51-61 poly (ADP-ribose) polymerase family, member 1 Mus musculus 26-30 33311600-0 2021 Anti-PD-L1 mediating tumor-targeted codelivery of liposomal irinotecan/JQ1 for chemo-immunotherapy. Irinotecan 60-70 CD274 molecule Homo sapiens 5-10 33311600-6 2021 Here, we develop a PD-L1-targeting immune liposome (P-Lipo) for co-delivering irinotecan (IRI) and JQ1, and this system can successfully elicit antitumor immunity in colorectal cancer through inducing ICD by IRI and interfering in the immunosuppressive PD-1/PD-L1 pathway by JQ1. Irinotecan 78-88 CD274 molecule Homo sapiens 19-24 33311600-6 2021 Here, we develop a PD-L1-targeting immune liposome (P-Lipo) for co-delivering irinotecan (IRI) and JQ1, and this system can successfully elicit antitumor immunity in colorectal cancer through inducing ICD by IRI and interfering in the immunosuppressive PD-1/PD-L1 pathway by JQ1. Irinotecan 78-88 programmed cell death 1 Homo sapiens 253-257 33311600-6 2021 Here, we develop a PD-L1-targeting immune liposome (P-Lipo) for co-delivering irinotecan (IRI) and JQ1, and this system can successfully elicit antitumor immunity in colorectal cancer through inducing ICD by IRI and interfering in the immunosuppressive PD-1/PD-L1 pathway by JQ1. Irinotecan 78-88 CD274 molecule Homo sapiens 258-263 33336394-0 2020 Cetuximab plus irinotecan administered biweekly with reduced infusion time to heavily pretreated patients with metastatic colorectal cancer and related RAS and BRAF mutation status. Irinotecan 15-25 B-Raf proto-oncogene, serine/threonine kinase Homo sapiens 160-164 33263997-1 2020 Tyrosyl-DNA phosphodiesterase 1 (TDP1) is a molecular target for the sensitization of cancer cells to the FDA-approved topoisomerase inhibitors topotecan and irinotecan. Irinotecan 158-168 tyrosyl-DNA phosphodiesterase 1 Homo sapiens 0-31 33263997-1 2020 Tyrosyl-DNA phosphodiesterase 1 (TDP1) is a molecular target for the sensitization of cancer cells to the FDA-approved topoisomerase inhibitors topotecan and irinotecan. Irinotecan 158-168 tyrosyl-DNA phosphodiesterase 1 Homo sapiens 33-37 33322698-6 2020 Chemosensitivity profiling of PDAC cell lines and patient-derived organoids found that CDKN2A inactivation was associated with the increased sensitivity to paclitaxel and SN-38 (the active metabolite of irinotecan). Irinotecan 171-176 cyclin dependent kinase inhibitor 2A Homo sapiens 87-93 33322698-6 2020 Chemosensitivity profiling of PDAC cell lines and patient-derived organoids found that CDKN2A inactivation was associated with the increased sensitivity to paclitaxel and SN-38 (the active metabolite of irinotecan). Irinotecan 203-213 cyclin dependent kinase inhibitor 2A Homo sapiens 87-93 33011217-11 2020 When combined with niclosamide or specific STAT3 inhibitor (C188-9), the cytotoxicity and DNA damage response from SN38 (the active metabolite from irinotecan) were significantly enhanced. Irinotecan 115-119 signal transducer and activator of transcription 3 Homo sapiens 43-48 33125947-0 2020 Pre-therapeutic UGT1A1 genotyping to reduce the risk of irinotecan-induced severe toxicity: Ready for prime time. Irinotecan 56-66 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 16-22 33125947-1 2020 BACKGROUND: Pre-therapeutic UGT1A1 genotyping is not yet routinely performed in most hospitals in patients starting irinotecan chemotherapy. Irinotecan 116-126 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 28-34 33125947-5 2020 A high level of evidence (level I) was found for a higher incidence of irinotecan-induced severe toxicity in homozygous carriers of UGT1A1*28 or UGT1A1*6. Irinotecan 71-81 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 132-138 33125947-5 2020 A high level of evidence (level I) was found for a higher incidence of irinotecan-induced severe toxicity in homozygous carriers of UGT1A1*28 or UGT1A1*6. Irinotecan 71-81 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 145-151 33125947-9 2020 CONCLUSION: Pre-therapeutic genotyping of UGT1A1 in patients initiating treatment with irinotecan improves patient safety, is likely to be cost-saving, and should, therefore, become standard of care. Irinotecan 87-97 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 42-48 33096057-4 2020 Moreover, the resistant cells suppressed formation of reactive 4-hydroxy-2-nonenal by CPT11 treatment, and the suppressive effect was almost completely abolished by addition of an AKR1C3 inhibitor. Irinotecan 86-91 aldo-keto reductase family 1 member C3 Homo sapiens 180-186 33011217-11 2020 When combined with niclosamide or specific STAT3 inhibitor (C188-9), the cytotoxicity and DNA damage response from SN38 (the active metabolite from irinotecan) were significantly enhanced. Irinotecan 148-158 signal transducer and activator of transcription 3 Homo sapiens 43-48 33152996-7 2020 Compared with the control condition, irinotecan diminished (p < 0.05) intestinal villus height, caused a loss of crypt integrity and intense inflammatory cell infiltration, increased myeloperoxidase (MPO), IL-6 and IL-1beta levels and decreased reduced glutathione (GSH) levels in duodenum segments and increased gastric retention and decreased liquid retention in the medial intestinal segment, resulting in increased intestinal transit, severe diarrhea and reduced survival (approximately 72%). Irinotecan 37-47 myeloperoxidase Mus musculus 183-198 33158067-6 2020 Mechanistic studies indicated that poziotinib increases the intracellular accumulation of the ABCB1 transporter substrates, paclitaxel and doxorubicin, and the ABCG2 transporter substrates, mitoxantrone and SN-38, by inhibiting their substrate efflux function. Irinotecan 207-212 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 160-165 33152996-7 2020 Compared with the control condition, irinotecan diminished (p < 0.05) intestinal villus height, caused a loss of crypt integrity and intense inflammatory cell infiltration, increased myeloperoxidase (MPO), IL-6 and IL-1beta levels and decreased reduced glutathione (GSH) levels in duodenum segments and increased gastric retention and decreased liquid retention in the medial intestinal segment, resulting in increased intestinal transit, severe diarrhea and reduced survival (approximately 72%). Irinotecan 37-47 myeloperoxidase Mus musculus 200-203 33152996-7 2020 Compared with the control condition, irinotecan diminished (p < 0.05) intestinal villus height, caused a loss of crypt integrity and intense inflammatory cell infiltration, increased myeloperoxidase (MPO), IL-6 and IL-1beta levels and decreased reduced glutathione (GSH) levels in duodenum segments and increased gastric retention and decreased liquid retention in the medial intestinal segment, resulting in increased intestinal transit, severe diarrhea and reduced survival (approximately 72%). Irinotecan 37-47 interleukin 6 Mus musculus 206-210 33152996-7 2020 Compared with the control condition, irinotecan diminished (p < 0.05) intestinal villus height, caused a loss of crypt integrity and intense inflammatory cell infiltration, increased myeloperoxidase (MPO), IL-6 and IL-1beta levels and decreased reduced glutathione (GSH) levels in duodenum segments and increased gastric retention and decreased liquid retention in the medial intestinal segment, resulting in increased intestinal transit, severe diarrhea and reduced survival (approximately 72%). Irinotecan 37-47 interleukin 1 alpha Mus musculus 215-223 33138058-13 2020 Both SN-38-incorporated micelles showed two-fold higher caspase-3/7 activation levels than irinotecan. Irinotecan 5-10 caspase 3 Homo sapiens 56-67 32863385-2 2020 METHODS: This Phase 2 trial evaluated the efficacy and safety of irinotecan plus cetuximab rechallenge as third-line treatment in KRAS wild-type mCRC patients who achieved clinical benefit with first-line cetuximab-containing therapy. Irinotecan 65-75 KRAS proto-oncogene, GTPase Homo sapiens 130-134 32863385-10 2020 CONCLUSIONS: Irinotecan plus cetuximab rechallenge as third-line treatment for KRAS wild-type mCRC was safe and had promising activity, especially in those with a long CFI, warranting further investigation in a Phase 3 randomised trial. Irinotecan 13-23 KRAS proto-oncogene, GTPase Homo sapiens 79-83 32863385-10 2020 CONCLUSIONS: Irinotecan plus cetuximab rechallenge as third-line treatment for KRAS wild-type mCRC was safe and had promising activity, especially in those with a long CFI, warranting further investigation in a Phase 3 randomised trial. Irinotecan 13-23 complement factor I Homo sapiens 168-171 32961616-2 2020 Here, we explore enhancement of the effect of irinotecan [camptothecin-11 (CPT-11)] by the p53-dependent induction of carboxylesterase 2 (CES2), a CPT-11-activating enzyme, in MM. Irinotecan 46-56 tumor protein p53 Homo sapiens 91-94 32961616-2 2020 Here, we explore enhancement of the effect of irinotecan [camptothecin-11 (CPT-11)] by the p53-dependent induction of carboxylesterase 2 (CES2), a CPT-11-activating enzyme, in MM. Irinotecan 46-56 carboxylesterase 2 Homo sapiens 118-136 32961616-2 2020 Here, we explore enhancement of the effect of irinotecan [camptothecin-11 (CPT-11)] by the p53-dependent induction of carboxylesterase 2 (CES2), a CPT-11-activating enzyme, in MM. Irinotecan 46-56 carboxylesterase 2 Homo sapiens 138-142 32961616-2 2020 Here, we explore enhancement of the effect of irinotecan [camptothecin-11 (CPT-11)] by the p53-dependent induction of carboxylesterase 2 (CES2), a CPT-11-activating enzyme, in MM. Irinotecan 58-73 tumor protein p53 Homo sapiens 91-94 32961616-2 2020 Here, we explore enhancement of the effect of irinotecan [camptothecin-11 (CPT-11)] by the p53-dependent induction of carboxylesterase 2 (CES2), a CPT-11-activating enzyme, in MM. Irinotecan 58-73 carboxylesterase 2 Homo sapiens 118-136 32961616-2 2020 Here, we explore enhancement of the effect of irinotecan [camptothecin-11 (CPT-11)] by the p53-dependent induction of carboxylesterase 2 (CES2), a CPT-11-activating enzyme, in MM. Irinotecan 58-73 carboxylesterase 2 Homo sapiens 138-142 32961616-2 2020 Here, we explore enhancement of the effect of irinotecan [camptothecin-11 (CPT-11)] by the p53-dependent induction of carboxylesterase 2 (CES2), a CPT-11-activating enzyme, in MM. Irinotecan 75-81 tumor protein p53 Homo sapiens 91-94 32961616-2 2020 Here, we explore enhancement of the effect of irinotecan [camptothecin-11 (CPT-11)] by the p53-dependent induction of carboxylesterase 2 (CES2), a CPT-11-activating enzyme, in MM. Irinotecan 75-81 carboxylesterase 2 Homo sapiens 118-136 32961616-2 2020 Here, we explore enhancement of the effect of irinotecan [camptothecin-11 (CPT-11)] by the p53-dependent induction of carboxylesterase 2 (CES2), a CPT-11-activating enzyme, in MM. Irinotecan 75-81 carboxylesterase 2 Homo sapiens 138-142 32961616-2 2020 Here, we explore enhancement of the effect of irinotecan [camptothecin-11 (CPT-11)] by the p53-dependent induction of carboxylesterase 2 (CES2), a CPT-11-activating enzyme, in MM. Irinotecan 147-153 tumor protein p53 Homo sapiens 91-94 32961616-2 2020 Here, we explore enhancement of the effect of irinotecan [camptothecin-11 (CPT-11)] by the p53-dependent induction of carboxylesterase 2 (CES2), a CPT-11-activating enzyme, in MM. Irinotecan 147-153 carboxylesterase 2 Homo sapiens 118-136 32961616-2 2020 Here, we explore enhancement of the effect of irinotecan [camptothecin-11 (CPT-11)] by the p53-dependent induction of carboxylesterase 2 (CES2), a CPT-11-activating enzyme, in MM. Irinotecan 147-153 carboxylesterase 2 Homo sapiens 138-142 32961616-5 2020 Knocking down CES2 in MM cells reduced the effect of the drug combination, and its forced expression in MESO4 cells enhanced the growth inhibitory activity of CPT-11 in the absence of nutlin-3a. Irinotecan 159-165 carboxylesterase 2 Homo sapiens 14-18 32737884-0 2020 Pharmacogenetic impact of UGT1A1 polymorphisms on pulmonary neuroendocrine tumours treated with metronomic irinotecan-based chemotherapy in Chinese populations. Irinotecan 107-117 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 26-32 32737884-11 2020 CONCLUSIONS: The impact of UGT1A1 polymorphisms varies in different irinotecan-based chemotherapies. Irinotecan 68-78 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 27-33 32737884-12 2020 UGT1A1*6 and UGTA1A*28 were useful for the prediction of irinotecan-related severe toxicity in Chinese PNT patients treated with metronomic IBC. Irinotecan 57-67 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 32936306-0 2020 Association of UGT1A1*6 polymorphism with irinotecan-based chemotherapy reaction in colorectal cancer patients: a systematic review and a meta-analysis. Irinotecan 42-52 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 15-21 32694423-11 2020 The biological function of anlotinib and irinotecan may be mediated through the AKT/ERK signaling pathway. Irinotecan 41-51 AKT serine/threonine kinase 1 Homo sapiens 80-83 32694423-11 2020 The biological function of anlotinib and irinotecan may be mediated through the AKT/ERK signaling pathway. Irinotecan 41-51 EPH receptor B2 Homo sapiens 84-87 32580836-7 2020 Moreover, the combined treatment of irinotecan with anti-hDLL4 or ABL001 showed synergistic antitumor activity. Irinotecan 36-46 delta like canonical Notch ligand 4 Homo sapiens 57-62 32580836-10 2020 Collectively, DLL4 inhibition significantly decreased cell motility and stem-like phenotype and the combination treatment of DLL4/VEGF bispecific therapeutic antibody with irinotecan synergistically reduced the GC burden in mouse models. Irinotecan 172-182 delta like canonical Notch ligand 4 Mus musculus 125-129 32580836-10 2020 Collectively, DLL4 inhibition significantly decreased cell motility and stem-like phenotype and the combination treatment of DLL4/VEGF bispecific therapeutic antibody with irinotecan synergistically reduced the GC burden in mouse models. Irinotecan 172-182 vascular endothelial growth factor A Mus musculus 130-134 33103961-6 2020 Conclusion: Therefore, the superparamagnetic iron oxide@poly(sodium styrene sulfonate)/irinotecan/human serum albumin-anti-CD133 nanoparticles are a powerful candidate for future antitumor strategies. Irinotecan 87-97 albumin Homo sapiens 104-117 33103961-6 2020 Conclusion: Therefore, the superparamagnetic iron oxide@poly(sodium styrene sulfonate)/irinotecan/human serum albumin-anti-CD133 nanoparticles are a powerful candidate for future antitumor strategies. Irinotecan 87-97 prominin 1 Homo sapiens 123-128 32985005-5 2020 In this review, the PGx profiles of drug-gene pairs with potential impact in GI malignancy therapy - DPYD-5-fluorouracil/capecitabine and UGT1A1-irinotecan - and the available clinical evidence of their roles in reducing severe adverse events are discussed. Irinotecan 145-155 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 138-144 33092063-2 2020 The activation of mechanistic Target Of Rapamycin (mTor)/hypoxia inducible factor (HIF)-1 pathway can be targeted by rapamycin and irinotecan, respectively. Irinotecan 131-141 mechanistic target of rapamycin kinase Homo sapiens 18-49 33120790-4 2020 PATIENT CONCERNS: A 72-year-old man diagnosed with stage IV rectal adenocarcinoma (KRAS mutation) with peritoneal carcinomatosis and liver metastases developed resistance to 2 lines of treatment (bevacizumab/irinotecan/S-1 and bevacizumab/oxaliplatin/HDFL [high-dose 24-hour infusion of 5-fluorouracil and leucovorin regimen]) within 5 months. Irinotecan 208-218 KRAS proto-oncogene, GTPase Homo sapiens 83-87 33092063-2 2020 The activation of mechanistic Target Of Rapamycin (mTor)/hypoxia inducible factor (HIF)-1 pathway can be targeted by rapamycin and irinotecan, respectively. Irinotecan 131-141 mechanistic target of rapamycin kinase Homo sapiens 51-55 33092063-2 2020 The activation of mechanistic Target Of Rapamycin (mTor)/hypoxia inducible factor (HIF)-1 pathway can be targeted by rapamycin and irinotecan, respectively. Irinotecan 131-141 hypoxia inducible factor 1 subunit alpha Homo sapiens 57-89 32829105-0 2020 Determination of the UGT1A1 polymorphism as guidance for irinotecan dose escalation in metastatic colorectal cancer treated with first-line bevacizumab and FOLFIRI (PURE FIST). Irinotecan 57-67 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 21-27 33069191-1 2021 OBJECTIVES: The aim of the present review is to discuss the potential link between RAS, BRAF and microsatellite instability (MSI) mutational patterns and chemotherapeutic agent efficacy [Irinotecan (IRI) vs. Oxaliplatin (OXA)], and how this can potentially influence the choice of the chemotherapy backbone. Irinotecan 187-197 B-Raf proto-oncogene, serine/threonine kinase Homo sapiens 88-92 32816496-0 2020 Treatment of Lung Cancer by Peptide-Modified Liposomal Irinotecan Endowed with Tumor Penetration and NF-kappaB Inhibitory Activities. Irinotecan 55-65 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 101-110 32816496-7 2020 These results strongly suggest that antitumor efficacy of the irinotecan liposomes can be enhanced by tumor-penetrating and NF-kappaB-inhibiting functions of CB5005. Irinotecan 62-72 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 124-133 32829105-1 2020 AIM: Uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) polymorphism plays a crucial role in the increased susceptibility of patients to irinotecan and its toxicity. Irinotecan 143-153 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 5-52 32829105-1 2020 AIM: Uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) polymorphism plays a crucial role in the increased susceptibility of patients to irinotecan and its toxicity. Irinotecan 143-153 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 54-60 32829105-7 2020 Dose escalation of irinotecan, an independent factor of ORR (P < 0.001) and DCR (P = 0.006), improved PFS in mCRC patients with wild-type and mutant KRAS (P = 0.007 and P = 0.019, respectively). Irinotecan 19-29 KRAS proto-oncogene, GTPase Homo sapiens 149-153 32829105-8 2020 CONCLUSION: The current study revealed that mCRC patients, regardless of KRAS gene status, with UGT1A1 genotyping can tolerate escalated doses of irinotecan and potentially achieve a more favourable clinical outcome without significantly increased toxicities. Irinotecan 146-156 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 96-102 32993166-0 2020 Injectable Thermo-Sensitive Chitosan Hydrogel Containing CPT-11-Loaded EGFR-Targeted Graphene Oxide and SLP2 shRNA for Localized Drug/Gene Delivery in Glioblastoma Therapy. Irinotecan 57-63 epidermal growth factor receptor Homo sapiens 71-75 32666389-0 2020 Impact of single-heterozygous UGT1A1 on the clinical outcomes of irinotecan monotherapy after fluoropyrimidine and platinum-based combination therapy for gastric cancer: a multicenter retrospective study. Irinotecan 65-75 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 30-36 32666389-2 2020 We investigated the association between clinical outcomes (efficacy and safety) and UGT1A1 status in patients who received irinotecan monotherapy. Irinotecan 123-133 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 84-90 32993166-2 2020 Toward this end, we load irinotecan (CPT-11) to cetuximab (CET)-conjugated GO (GO-CET/CPT11) for pH-responsive drug release after endocytosis by epidermal growth factor receptor (EGFR) over-expressed U87 human glioblastoma cells. Irinotecan 25-35 epidermal growth factor receptor Homo sapiens 145-177 32993166-2 2020 Toward this end, we load irinotecan (CPT-11) to cetuximab (CET)-conjugated GO (GO-CET/CPT11) for pH-responsive drug release after endocytosis by epidermal growth factor receptor (EGFR) over-expressed U87 human glioblastoma cells. Irinotecan 25-35 epidermal growth factor receptor Homo sapiens 179-183 32993166-2 2020 Toward this end, we load irinotecan (CPT-11) to cetuximab (CET)-conjugated GO (GO-CET/CPT11) for pH-responsive drug release after endocytosis by epidermal growth factor receptor (EGFR) over-expressed U87 human glioblastoma cells. Irinotecan 37-43 epidermal growth factor receptor Homo sapiens 145-177 32993166-2 2020 Toward this end, we load irinotecan (CPT-11) to cetuximab (CET)-conjugated GO (GO-CET/CPT11) for pH-responsive drug release after endocytosis by epidermal growth factor receptor (EGFR) over-expressed U87 human glioblastoma cells. Irinotecan 37-43 epidermal growth factor receptor Homo sapiens 179-183 32993166-6 2020 Confocal microscopy analysis confirmed the intracellular trafficking for the targeted delivery of CPT-11 through interactions of CET with EGFR on the U87 cell surface. Irinotecan 98-104 epidermal growth factor receptor Homo sapiens 138-142 33042471-4 2020 The biological significance of CD44 expression in the chemoresistance response to fluorouracil, oxaliplatin or irinotecan, three major anti-cancer agents for colon cancer in the clinical setting, was examined using colon cancer cell lines. Irinotecan 111-121 CD44 molecule (Indian blood group) Homo sapiens 31-35 33042265-5 2020 Methods: Using a microfluidic microbubble production platform, we generated thMBs comprising VEGFR2-targeted microbubbles with attached liposomal payloads for localised ultrasound-triggered delivery of irinotecan and SN38 in mouse models of colorectal cancer. Irinotecan 202-212 kinase insert domain protein receptor Mus musculus 93-99 32927892-2 2020 It is well-known that irinotecan as a chemotherapeutic drug against non-small-cell lung carcinoma (NSCLC) has limited anticancer effect due to NF-kappaB activation. Irinotecan 22-32 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 143-152 32927892-12 2020 Collectively, irinotecan and GRA16 co-treatment promotes the anticancer effects of irinotecan via NF-kappaB inhibition and cell cycle arrest induced by GRA16, subsequently increasing the chemotherapeutic effect of irinotecan to NSCLC cells via NF-kappaB inhibition. Irinotecan 14-24 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 98-107 32927892-12 2020 Collectively, irinotecan and GRA16 co-treatment promotes the anticancer effects of irinotecan via NF-kappaB inhibition and cell cycle arrest induced by GRA16, subsequently increasing the chemotherapeutic effect of irinotecan to NSCLC cells via NF-kappaB inhibition. Irinotecan 14-24 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 244-253 32927892-12 2020 Collectively, irinotecan and GRA16 co-treatment promotes the anticancer effects of irinotecan via NF-kappaB inhibition and cell cycle arrest induced by GRA16, subsequently increasing the chemotherapeutic effect of irinotecan to NSCLC cells via NF-kappaB inhibition. Irinotecan 83-93 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 98-107 32927892-12 2020 Collectively, irinotecan and GRA16 co-treatment promotes the anticancer effects of irinotecan via NF-kappaB inhibition and cell cycle arrest induced by GRA16, subsequently increasing the chemotherapeutic effect of irinotecan to NSCLC cells via NF-kappaB inhibition. Irinotecan 83-93 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 244-253 32927892-12 2020 Collectively, irinotecan and GRA16 co-treatment promotes the anticancer effects of irinotecan via NF-kappaB inhibition and cell cycle arrest induced by GRA16, subsequently increasing the chemotherapeutic effect of irinotecan to NSCLC cells via NF-kappaB inhibition. Irinotecan 83-93 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 98-107 32927892-12 2020 Collectively, irinotecan and GRA16 co-treatment promotes the anticancer effects of irinotecan via NF-kappaB inhibition and cell cycle arrest induced by GRA16, subsequently increasing the chemotherapeutic effect of irinotecan to NSCLC cells via NF-kappaB inhibition. Irinotecan 83-93 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 244-253 32548889-1 2020 BACKGROUND AND PURPOSE: Irinotecan, a drug used in colorectal cancer therapy is metabolized by glucuronidation involving different UDP-glucuronosyltransferase (UGT)1A isoforms, which leads to facilitated elimination from the body. Irinotecan 24-34 Ugt1a@ Mus musculus 160-166 32548889-2 2020 Individuals homozygous for the genetic variants UGT1A1*28 (Gilbert syndrome) and UGT1A7*3 are more susceptible to irinotecan drug side effects such as severe diarrhea and leukopenia. Irinotecan 114-124 UDP glucuronosyltransferase 1 family, polypeptide A1 Mus musculus 48-54 32548889-2 2020 Individuals homozygous for the genetic variants UGT1A1*28 (Gilbert syndrome) and UGT1A7*3 are more susceptible to irinotecan drug side effects such as severe diarrhea and leukopenia. Irinotecan 114-124 UDP glucuronosyltransferase 1 family, polypeptide A6A Mus musculus 81-87 32548889-6 2020 KEY RESULTS: Only the combination of the two coffee ingredients, CA and CAPE, mediates the protective effects of coffee in a model of irinotecan toxicity by activation of UGT1A genes. Irinotecan 134-144 Ugt1a@ Mus musculus 171-176 32532788-0 2020 Plasma Interleukin-8 is a biomarker for TAK1 activation and predicts resistance to nanoliposomal irinotecan in patients with gemcitabine-refractory pancreatic cancer. Irinotecan 97-107 C-X-C motif chemokine ligand 8 Homo sapiens 7-20 32846370-7 2020 ATRX mutant cells also showed selective sensitivity to the DNA damaging agents, sapacitabine and irinotecan. Irinotecan 97-107 ATRX chromatin remodeler Homo sapiens 0-4 32846370-8 2020 HRR deficiency was also seen in the ATRX deleted CHLA-90 cell line, and significant sensitivity demonstrated to olaparib/irinotecan combination therapy in all ATRX LoF models. Irinotecan 121-131 ATRX chromatin remodeler Homo sapiens 159-163 32846370-9 2020 In-vivo sensitivity to olaparib/irinotecan was seen in ATRX mutant but not wild-type xenografts. Irinotecan 32-42 ATRX chromatin remodeler Homo sapiens 55-59 32846370-10 2020 Finally, sustained responses to olaparib/irinotecan therapy were seen in an ATRX deleted neuroblastoma patient derived xenograft. Irinotecan 41-51 ATRX chromatin remodeler Homo sapiens 76-80 32846370-12 2020 In ATRX LoF models, preclinical sensitivity is demonstrated to olaparib and irinotecan, a combination that can be rapidly translated into the clinic. Irinotecan 76-86 ATRX chromatin remodeler Homo sapiens 3-7 32776374-6 2020 CPT-11 increased levels of tissue factor (TF) both in the blood and in intestines, and decreased the intestinal blood flow in mice. Irinotecan 0-6 coagulation factor III Mus musculus 27-40 32776374-6 2020 CPT-11 increased levels of tissue factor (TF) both in the blood and in intestines, and decreased the intestinal blood flow in mice. Irinotecan 0-6 coagulation factor III Mus musculus 42-44 32776374-10 2020 Ozone inhibited TF expression induced by CPT-11 via activating AMPK/SOCS3, and effectively ameliorated the intestinal mucosal injury in mice. Irinotecan 41-47 coagulation factor III Mus musculus 16-18 32776374-10 2020 Ozone inhibited TF expression induced by CPT-11 via activating AMPK/SOCS3, and effectively ameliorated the intestinal mucosal injury in mice. Irinotecan 41-47 suppressor of cytokine signaling 3 Mus musculus 68-73 32502622-0 2020 P53 activation suppresses irinotecan metabolite SN-38-induced cell damage in non-malignant but not malignant epithelial colonic cells. Irinotecan 26-36 tumor protein p53 Homo sapiens 0-3 32502622-0 2020 P53 activation suppresses irinotecan metabolite SN-38-induced cell damage in non-malignant but not malignant epithelial colonic cells. Irinotecan 48-53 tumor protein p53 Homo sapiens 0-3 32502622-5 2020 SN-38 treatment induced significant apoptosis in all cell lines after 48 h. Nutlin-3a unexpectedly increased cell death in the p53 wild-type CRC cell line, HCT116+/+, while Nutlin-3a pre-treatment provided protection from SN-38 in the p53 wild-type normal cell line, FHS 74. Irinotecan 0-5 tumor protein p53 Homo sapiens 127-130 32502622-5 2020 SN-38 treatment induced significant apoptosis in all cell lines after 48 h. Nutlin-3a unexpectedly increased cell death in the p53 wild-type CRC cell line, HCT116+/+, while Nutlin-3a pre-treatment provided protection from SN-38 in the p53 wild-type normal cell line, FHS 74. Irinotecan 0-5 tumor protein p53 Homo sapiens 235-238 32778670-3 2020 Thus, the aim of this study was to assess the allele and genotype frequencies and the relationship between CYP3A4/5, DPYD, UGT1A1, ABCB1, and ABCC2 genetic variations and irinotecan-induced toxicity in Thai colorectal cancer patients. Irinotecan 171-181 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 107-115 32778670-10 2020 This finding suggests that UGT1A1*28, *6, and ABCC2 c.3972C > T might be an important predictor for irinotecan-induced severe neutropenia. Irinotecan 100-110 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 27-33 32778670-3 2020 Thus, the aim of this study was to assess the allele and genotype frequencies and the relationship between CYP3A4/5, DPYD, UGT1A1, ABCB1, and ABCC2 genetic variations and irinotecan-induced toxicity in Thai colorectal cancer patients. Irinotecan 171-181 dihydropyrimidine dehydrogenase Homo sapiens 117-121 32778670-10 2020 This finding suggests that UGT1A1*28, *6, and ABCC2 c.3972C > T might be an important predictor for irinotecan-induced severe neutropenia. Irinotecan 100-110 ATP binding cassette subfamily C member 2 Homo sapiens 46-51 32778670-3 2020 Thus, the aim of this study was to assess the allele and genotype frequencies and the relationship between CYP3A4/5, DPYD, UGT1A1, ABCB1, and ABCC2 genetic variations and irinotecan-induced toxicity in Thai colorectal cancer patients. Irinotecan 171-181 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 123-129 32821126-8 2020 Results: The expression of MCT1 and LDHB was high in the liver metastases of irinotecan-resistant patients, and the high level of IL-6 in the plasma of patients with CRLM was associated with poor response to irinotecan-based chemotherapy. Irinotecan 77-87 solute carrier family 16 member 1 Homo sapiens 27-31 32764831-0 2020 CTDSP1 inhibitor rabeprazole regulates DNA-PKcs dependent topoisomerase I degradation and irinotecan drug resistance in colorectal cancer. Irinotecan 90-100 CTD small phosphatase 1 Homo sapiens 0-6 32764831-3 2020 We have published that the deregulated DNA-PKcs kinase cascade ensures rapid degradation of topoI and is at the core of the drug resistance mechanism of topoI inhibitors, including irinotecan. Irinotecan 181-191 protein kinase, DNA-activated, catalytic subunit Homo sapiens 39-47 32764831-7 2020 Using differentially expressing CTDSP1 cells, we demonstrated that CTDSP1 contributes to the irinotecan sensitivity by preventing topoI degradation. Irinotecan 93-103 CTD small phosphatase 1 Homo sapiens 32-38 32764831-7 2020 Using differentially expressing CTDSP1 cells, we demonstrated that CTDSP1 contributes to the irinotecan sensitivity by preventing topoI degradation. Irinotecan 93-103 CTD small phosphatase 1 Homo sapiens 67-73 32764831-8 2020 Retrospective analysis of patients receiving irinotecan with or without rabeprazole has shown the effects of CTDSP1 on irinotecan response. Irinotecan 45-55 CTD small phosphatase 1 Homo sapiens 109-115 32764831-8 2020 Retrospective analysis of patients receiving irinotecan with or without rabeprazole has shown the effects of CTDSP1 on irinotecan response. Irinotecan 119-129 CTD small phosphatase 1 Homo sapiens 109-115 32764831-9 2020 These results indicate that CTDSP1 promotes sensitivity to irinotecan and rabeprazole prevents this effect, resulting in drug resistance. Irinotecan 59-69 CTD small phosphatase 1 Homo sapiens 28-34 32821126-8 2020 Results: The expression of MCT1 and LDHB was high in the liver metastases of irinotecan-resistant patients, and the high level of IL-6 in the plasma of patients with CRLM was associated with poor response to irinotecan-based chemotherapy. Irinotecan 77-87 lactate dehydrogenase B Homo sapiens 36-40 32821126-8 2020 Results: The expression of MCT1 and LDHB was high in the liver metastases of irinotecan-resistant patients, and the high level of IL-6 in the plasma of patients with CRLM was associated with poor response to irinotecan-based chemotherapy. Irinotecan 208-218 solute carrier family 16 member 1 Homo sapiens 27-31 32821126-8 2020 Results: The expression of MCT1 and LDHB was high in the liver metastases of irinotecan-resistant patients, and the high level of IL-6 in the plasma of patients with CRLM was associated with poor response to irinotecan-based chemotherapy. Irinotecan 208-218 interleukin 6 Homo sapiens 130-134 32529410-1 2020 Sacituzumab govitecan (sacituzumab govitecan-hziy; Trodelvy ) is a Trop-2-directed antibody conjugated to a topoisomerase I inhibitor (SN-38) that is being developed by Immunomedics for the treatment of solid tumours, including breast cancer. Irinotecan 135-140 tumor associated calcium signal transducer 2 Homo sapiens 67-73 32758288-0 2020 UGT1A1 polymorphism has a prognostic effect in patients with stage IB or II uterine cervical cancer and one or no metastatic pelvic nodes receiving irinotecan chemotherapy: a retrospective study. Irinotecan 148-158 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 32758288-1 2020 BACKGROUND: Uridine diphosphate glucuronosyltransferase 1 family polypeptide A1 (UGT1A1) is a predictive biomarker for the side-effects of irinotecan chemotherapy, which reduces the volume of tumors harboring UGT1A1 polymorphisms. Irinotecan 139-149 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-79 32758288-1 2020 BACKGROUND: Uridine diphosphate glucuronosyltransferase 1 family polypeptide A1 (UGT1A1) is a predictive biomarker for the side-effects of irinotecan chemotherapy, which reduces the volume of tumors harboring UGT1A1 polymorphisms. Irinotecan 139-149 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 81-87 32758288-1 2020 BACKGROUND: Uridine diphosphate glucuronosyltransferase 1 family polypeptide A1 (UGT1A1) is a predictive biomarker for the side-effects of irinotecan chemotherapy, which reduces the volume of tumors harboring UGT1A1 polymorphisms. Irinotecan 139-149 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 209-215 32758288-2 2020 We aimed to determine whether UGT1A1 polymorphisms can predict progression-free survival in patients with local cervical cancer treated with irinotecan chemotherapy. Irinotecan 141-151 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 30-36 32758288-10 2020 Among patients with <=1 metastatic lymph node, those with UGT1A1 polymorphisms benefited from irinotecan chemotherapy and demonstrated significantly longer progression-free survival (p = 0.020) than those with wild-type UGT1A1. Irinotecan 94-104 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 58-64 32758288-11 2020 CONCLUSIONS: Irinotecan chemotherapy might be beneficial in patients with cervical cancer, UGT1A1 polymorphisms, and <= 1 metastatic lymph nodes. Irinotecan 13-23 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 91-97 32850329-8 2020 Pharmacogenomic analyses revealed multiple alterations of fluoropyrimidine and irinotecan metabolism: severe deficiency of fluorouracil degradation rate (FUDR), single nucleotide polymorphisms of UGT1A1 *28 variable number of tandem repeats (VNTR) 7R/7R homozygote, ABCB1 c.C3435T, c.C1236T, MTHFR c.C667T homozygote, DPYD c.A166G, TSER 28bp VNTR 2R/3R heterozygote. Irinotecan 79-89 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 196-202 32387453-10 2020 Patients with right-PTL and high EREG/AREG or HER3 expression, had IrPan PFS improvement (high EREG/AREG HR=0.20, p=0.04; high HER3 HR=0.33, p=0.10) compared with irinotecan. Irinotecan 163-173 epiregulin Homo sapiens 33-37 32387453-10 2020 Patients with right-PTL and high EREG/AREG or HER3 expression, had IrPan PFS improvement (high EREG/AREG HR=0.20, p=0.04; high HER3 HR=0.33, p=0.10) compared with irinotecan. Irinotecan 163-173 amphiregulin Homo sapiens 38-42 32487736-9 2020 Interestingly, irinotecan selectively inhibited hOCT1 whereas crizotinib potently inhibited hOCT3-mediated mIBG uptake. Irinotecan 15-25 solute carrier family 22 member 1 Homo sapiens 48-53 34007623-0 2020 Dosage Adjustment of Irinotecan in Patients with UGT1A1 Polymorphisms: A Review of Current Literature. Irinotecan 21-31 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 49-55 34007623-1 2020 Objective: To review available literature regarding pharmacogenomics (PGx) effects on the metabolism of irinotecan by the UGT1A1 gene and the resulting dose adjustments based on PGx genetic variant. Irinotecan 104-114 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 122-128 34007623-3 2020 The extent of decreased function of UGT1A1 varies based on genotype so irinotecan dose adjustments may be needed. Irinotecan 71-81 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 36-42 34007623-7 2020 Conclusion: These findings add to a growing body of literature that suggest patients with UGT1A1 *28 or *6 variant alleles benefit from lower doses of irinotecan. Irinotecan 151-161 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 90-96 32708825-0 2020 ABCG2 Protein Levels and Association to Response to First-Line Irinotecan-Based Therapy for Patients with Metastatic Colorectal Cancer. Irinotecan 63-73 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 0-5 32708825-1 2020 In this study we investigated the use of cancer cell protein expression of ABCG2 to predict efficacy of systemic first-line irinotecan containing therapy in patients with metastatic colorectal cancer (mCRC). Irinotecan 124-134 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 75-80 32708825-10 2020 These results suggest that measurement of the ABCG2 drug efflux pump might be used to select patients with mCRC for irinotecan treatment. Irinotecan 116-126 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 46-51 32708825-12 2020 Moreover, patients with high ABCG2 immunoreactivity could be candidates for specific ABCG2 inhibition treatment in combination with irinotecan. Irinotecan 132-142 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 29-34 32503654-4 2020 Therefore, this trial was designed to observe whether irinotecan with a ketogenic diet can promote sensitivity to chemotherapy and remit target lesions in locally recurrent or metastatic Her-2-negative breast cancer patients. Irinotecan 54-64 erb-b2 receptor tyrosine kinase 2 Homo sapiens 187-192 32387182-0 2020 Irinotecan-mediated diarrhea is mainly correlated with intestinal exposure to SN-38: Critical role of gut Ugt. Irinotecan 0-10 solute carrier family 35 (UDP-galactose transporter), member A2 Mus musculus 106-109 32387182-13 2020 CONCLUSION AND IMPLICATIONS: Intestinal SN-38 exposure is mainly affected by intestinal Ugt activities and blood esterase activities, and strongly correlated with severity of IID. Irinotecan 40-45 solute carrier family 35 (UDP-galactose transporter), member A2 Mus musculus 88-91 32695000-0 2020 ABCB1 Genetic Variants as Predictors of Irinotecan-Induced Severe Gastrointestinal Toxicity in Metastatic Colorectal Cancer Patients. Irinotecan 40-50 ATP binding cassette subfamily B member 1 Homo sapiens 0-5 32695000-5 2020 P-gp is involved in the biliary excretion of irinotecan and its active metabolite SN-38. Irinotecan 45-55 ATP binding cassette subfamily B member 1 Homo sapiens 0-4 32695000-5 2020 P-gp is involved in the biliary excretion of irinotecan and its active metabolite SN-38. Irinotecan 82-87 ATP binding cassette subfamily B member 1 Homo sapiens 0-4 32184294-9 2020 Pharmacodynamic markers induced by eribulin (phospho-histone H3) and irinotecan (gamma-H2A.X) were abrogated in combination treated tumors. Irinotecan 69-79 H2A.X variant histone Mus musculus 81-92 32184294-16 2020 Although a mechanism for in vivo synergy requires further study, it is possible that eribulin-induced inhibition of microtubule dynamics enhances irinotecan-induced nuclear accumulation of TP53, leading to rapid cell death. Irinotecan 146-156 transformation related protein 53 Mus musculus 189-193 32343642-0 2020 Irinotecan, Temozolomide, and Dinutuximab With GM-CSF in Children With Refractory or Relapsed Neuroblastoma: A Report From the Children"s Oncology Group. Irinotecan 0-10 colony stimulating factor 2 Homo sapiens 47-53 31479512-7 2020 Based on concurrent biomarker expression, combinations of immunotherapy with platinum (ERCC1 negativity) or with doxorubicin, epirubicin, or etoposide (TOP2A positivity), have a higher probability of response while combinations with irinotecan or topotecan (TOPO1 positivity), with gemcitabine (RRM1 negativity), and fluorouracil, pemetrexed, or capecitabine (TS negativity) may be of less benefit. Irinotecan 233-243 ERCC excision repair 1, endonuclease non-catalytic subunit Homo sapiens 87-92 32265117-12 2020 CONCLUSION: Our analysis suggested higher completion of concurrent irinotecan was associated with better tumor response for patients with locally advanced rectal cancer with UGT1A1*1*1 or UGT1A1*1*28 phenotypes in the neoadjuvant setting, and at least 4 cycles was recommended. Irinotecan 67-77 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 174-180 32265117-12 2020 CONCLUSION: Our analysis suggested higher completion of concurrent irinotecan was associated with better tumor response for patients with locally advanced rectal cancer with UGT1A1*1*1 or UGT1A1*1*28 phenotypes in the neoadjuvant setting, and at least 4 cycles was recommended. Irinotecan 67-77 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 188-194 31479512-7 2020 Based on concurrent biomarker expression, combinations of immunotherapy with platinum (ERCC1 negativity) or with doxorubicin, epirubicin, or etoposide (TOP2A positivity), have a higher probability of response while combinations with irinotecan or topotecan (TOPO1 positivity), with gemcitabine (RRM1 negativity), and fluorouracil, pemetrexed, or capecitabine (TS negativity) may be of less benefit. Irinotecan 233-243 DNA topoisomerase II alpha Homo sapiens 152-157 32084420-6 2020 Interestingly, p53 expression was elevated by the SN-38 mono-treatment, and was not further increased after the co-treatment; besides, knockdown of p53 could only reduce the expression of cleaved-PARP partially, and weaken the enhanced proliferation inhibition induced by SN-38 plus imatinib, indicating that there might be other factors involved in the synergistic effects besides p53. Irinotecan 50-55 tumor protein p53 Homo sapiens 15-18 32471996-0 2020 Soluble HLA-G expression levels and HLA-G/irinotecan association in metastatic colorectal cancer treated with irinotecan-based strategy. Irinotecan 110-120 major histocompatibility complex, class I, G Homo sapiens 8-13 32471996-3 2020 The results suggest a link between HLA-G levels and irinotecan (CPT-11) pharmacokinetic, leading to hypothesize a molecular interaction between sHLA-G and CPT-11. Irinotecan 52-62 major histocompatibility complex, class I, G Homo sapiens 35-40 32471996-3 2020 The results suggest a link between HLA-G levels and irinotecan (CPT-11) pharmacokinetic, leading to hypothesize a molecular interaction between sHLA-G and CPT-11. Irinotecan 64-70 major histocompatibility complex, class I, G Homo sapiens 35-40 32471996-3 2020 The results suggest a link between HLA-G levels and irinotecan (CPT-11) pharmacokinetic, leading to hypothesize a molecular interaction between sHLA-G and CPT-11. Irinotecan 155-161 major histocompatibility complex, class I, G Homo sapiens 35-40 32471996-6 2020 The interaction between HLA-G polymorphs and CPT-11 was explored by means of computational modelling, confirming the hypothesis that CPT-11 could actually target the peptide binding cleft of the most common HLA-G polymorphs. Irinotecan 45-51 major histocompatibility complex, class I, G Homo sapiens 24-29 32471996-6 2020 The interaction between HLA-G polymorphs and CPT-11 was explored by means of computational modelling, confirming the hypothesis that CPT-11 could actually target the peptide binding cleft of the most common HLA-G polymorphs. Irinotecan 45-51 major histocompatibility complex, class I, G Homo sapiens 207-212 32084420-6 2020 Interestingly, p53 expression was elevated by the SN-38 mono-treatment, and was not further increased after the co-treatment; besides, knockdown of p53 could only reduce the expression of cleaved-PARP partially, and weaken the enhanced proliferation inhibition induced by SN-38 plus imatinib, indicating that there might be other factors involved in the synergistic effects besides p53. Irinotecan 50-55 poly(ADP-ribose) polymerase 1 Homo sapiens 196-200 32084420-6 2020 Interestingly, p53 expression was elevated by the SN-38 mono-treatment, and was not further increased after the co-treatment; besides, knockdown of p53 could only reduce the expression of cleaved-PARP partially, and weaken the enhanced proliferation inhibition induced by SN-38 plus imatinib, indicating that there might be other factors involved in the synergistic effects besides p53. Irinotecan 272-277 tumor protein p53 Homo sapiens 15-18 32084420-6 2020 Interestingly, p53 expression was elevated by the SN-38 mono-treatment, and was not further increased after the co-treatment; besides, knockdown of p53 could only reduce the expression of cleaved-PARP partially, and weaken the enhanced proliferation inhibition induced by SN-38 plus imatinib, indicating that there might be other factors involved in the synergistic effects besides p53. Irinotecan 272-277 tumor protein p53 Homo sapiens 148-151 32084420-6 2020 Interestingly, p53 expression was elevated by the SN-38 mono-treatment, and was not further increased after the co-treatment; besides, knockdown of p53 could only reduce the expression of cleaved-PARP partially, and weaken the enhanced proliferation inhibition induced by SN-38 plus imatinib, indicating that there might be other factors involved in the synergistic effects besides p53. Irinotecan 272-277 poly(ADP-ribose) polymerase 1 Homo sapiens 196-200 32084420-6 2020 Interestingly, p53 expression was elevated by the SN-38 mono-treatment, and was not further increased after the co-treatment; besides, knockdown of p53 could only reduce the expression of cleaved-PARP partially, and weaken the enhanced proliferation inhibition induced by SN-38 plus imatinib, indicating that there might be other factors involved in the synergistic effects besides p53. Irinotecan 272-277 tumor protein p53 Homo sapiens 148-151 32393425-0 2020 Effects of cyclosporin A on the pharmacokinetics and toxicities of irinotecan mediated by UGT1A1 in vitro and in vivo. Irinotecan 67-77 UDP glucuronosyltransferase family 1 member A1 Rattus norvegicus 90-96 32393425-4 2020 At present, most studies focused on the inhibition of bile excretion by cyclosporin A through the transporters MRP2 and MDR1 and its effect the irinotecan treatment in vivo. Irinotecan 144-154 ATP binding cassette subfamily C member 2 Rattus norvegicus 111-115 32393425-4 2020 At present, most studies focused on the inhibition of bile excretion by cyclosporin A through the transporters MRP2 and MDR1 and its effect the irinotecan treatment in vivo. Irinotecan 144-154 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 120-124 32393425-5 2020 However, UDP glucuronyltransferase-1 polypeptide A1 (UGT1A1) was related to a significantly altered disposition of irinotecan and its metabolites, and was therefore associated with irinotecan-induced toxicity. Irinotecan 115-125 UDP glucuronosyltransferase family 1 member A1 Rattus norvegicus 9-51 32393425-5 2020 However, UDP glucuronyltransferase-1 polypeptide A1 (UGT1A1) was related to a significantly altered disposition of irinotecan and its metabolites, and was therefore associated with irinotecan-induced toxicity. Irinotecan 115-125 UDP glucuronosyltransferase family 1 member A1 Rattus norvegicus 53-59 32393425-5 2020 However, UDP glucuronyltransferase-1 polypeptide A1 (UGT1A1) was related to a significantly altered disposition of irinotecan and its metabolites, and was therefore associated with irinotecan-induced toxicity. Irinotecan 181-191 UDP glucuronosyltransferase family 1 member A1 Rattus norvegicus 9-51 32393425-5 2020 However, UDP glucuronyltransferase-1 polypeptide A1 (UGT1A1) was related to a significantly altered disposition of irinotecan and its metabolites, and was therefore associated with irinotecan-induced toxicity. Irinotecan 181-191 UDP glucuronosyltransferase family 1 member A1 Rattus norvegicus 53-59 32393425-6 2020 This study focused on UGT1A1-mediated conversion of SN-38 to SN-38G, and systematically investigated the CsA-irinotecan interactions in vitro and in vivo. Irinotecan 52-57 UDP glucuronosyltransferase family 1 member A1 Rattus norvegicus 22-28 32393425-6 2020 This study focused on UGT1A1-mediated conversion of SN-38 to SN-38G, and systematically investigated the CsA-irinotecan interactions in vitro and in vivo. Irinotecan 61-66 UDP glucuronosyltransferase family 1 member A1 Rattus norvegicus 22-28 32393425-11 2020 The present study indicated that CsA treatment could enhance the systemic exposure and toxicity of SN-38 by inhibiting the UGT1A1 enzyme. Irinotecan 99-104 UDP glucuronosyltransferase family 1 member A1 Rattus norvegicus 123-129 32393425-12 2020 The inhibition of UGT1A1 enzyme might be a critical factor in the failure of CsA improving irinotecan"s treatment index. Irinotecan 91-101 UDP glucuronosyltransferase family 1 member A1 Rattus norvegicus 18-24 32264830-0 2020 Correlation between UGT1A1 gene polymorphism and irinotecan chemotherapy in metastatic colorectal cancer: a study from Guangxi Zhuang. Irinotecan 49-59 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 20-26 32349240-7 2020 Dual inhibition of P-gp/Bcrp, or Mrp showed a significant increase on SN-38 BBB transport: Cerebrum (8.3-fold and 3-fold, respectively), cerebellum (4.2-fold and 2.8-fold), and brainstem (2.6-fold and 2.2-fold). Irinotecan 70-75 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 19-23 32349240-7 2020 Dual inhibition of P-gp/Bcrp, or Mrp showed a significant increase on SN-38 BBB transport: Cerebrum (8.3-fold and 3-fold, respectively), cerebellum (4.2-fold and 2.8-fold), and brainstem (2.6-fold and 2.2-fold). Irinotecan 70-75 ATP binding cassette subfamily C member 1 Rattus norvegicus 33-36 32326511-2 2020 We tested ABCG2 and topoisomerase 1 (TOP1) mRNA expression as predictive biomarkers for irinotecan benefit in the PETACC-3 patient cohort. Irinotecan 88-98 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 10-15 32264830-14 2020 The UGT1A1 gene polymorphism with irinotecan chemotherapy-associated diarrhea and neutropenia were closely related. Irinotecan 34-44 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 32308415-0 2020 Irinotecan Induces Autophagy-Dependent Apoptosis and Positively Regulates ROS-Related JNK- and P38-MAPK Pathways in Gastric Cancer Cells. Irinotecan 0-10 mitogen-activated protein kinase 8 Homo sapiens 86-89 32308415-0 2020 Irinotecan Induces Autophagy-Dependent Apoptosis and Positively Regulates ROS-Related JNK- and P38-MAPK Pathways in Gastric Cancer Cells. Irinotecan 0-10 mitogen-activated protein kinase 14 Homo sapiens 95-103 32308415-9 2020 A mechanistic analysis showed that IRI-induced autophagy and apoptosis were related to increased reactive oxygen species (ROS) accumulation and activation of the JNK- and p38-MAPK pathways. Irinotecan 35-38 mitogen-activated protein kinase 8 Homo sapiens 162-165 32308415-9 2020 A mechanistic analysis showed that IRI-induced autophagy and apoptosis were related to increased reactive oxygen species (ROS) accumulation and activation of the JNK- and p38-MAPK pathways. Irinotecan 35-38 mitogen-activated protein kinase 14 Homo sapiens 171-179 30514181-4 2020 Irinotecan is bioactivated into its metabolite SN-38, which is subsequently detoxified by uridine diphosphate-glucuronosyl transferases (mainly UGT1A1). Irinotecan 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 144-150 31924737-12 2020 CONCLUSIONS: Tolerability and activity observed in this phase I trial support further investigation of the FTD/TPI-irinotecan-bevacizumab combination in previously treated mCRC. Irinotecan 115-125 triosephosphate isomerase 1 Homo sapiens 111-114 31975441-4 2020 BCRP and P-gp functions were evaluated by means of efflux and uptake assays using 7-ethyl-10-hydroxycamptothecin (SN-38) and rhodamine123 (Rho123) as specific substrates, respectively, in non-small cell lung cancer HCC827 cells, intestinal cancer Caco-2 cells and renal cancer Caki-1 cells. Irinotecan 82-112 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 0-4 31975441-4 2020 BCRP and P-gp functions were evaluated by means of efflux and uptake assays using 7-ethyl-10-hydroxycamptothecin (SN-38) and rhodamine123 (Rho123) as specific substrates, respectively, in non-small cell lung cancer HCC827 cells, intestinal cancer Caco-2 cells and renal cancer Caki-1 cells. Irinotecan 82-112 phosphoglycolate phosphatase Homo sapiens 9-13 31975441-4 2020 BCRP and P-gp functions were evaluated by means of efflux and uptake assays using 7-ethyl-10-hydroxycamptothecin (SN-38) and rhodamine123 (Rho123) as specific substrates, respectively, in non-small cell lung cancer HCC827 cells, intestinal cancer Caco-2 cells and renal cancer Caki-1 cells. Irinotecan 114-119 phosphoglycolate phosphatase Homo sapiens 9-13 31975441-5 2020 KEY FINDINGS: In HCC827 cells, the efflux rates of SN-38 and Rho123 were significantly decreased by knockdown of Ezr or Msn, but not Rdx. Irinotecan 51-56 ezrin Homo sapiens 113-116 31975441-5 2020 KEY FINDINGS: In HCC827 cells, the efflux rates of SN-38 and Rho123 were significantly decreased by knockdown of Ezr or Msn, but not Rdx. Irinotecan 51-56 moesin Homo sapiens 120-123 31975441-7 2020 In Caki-1 cells, Rdx knockdown increased the intracellular SN-38 concentration, while knockdown of Ezr or Msn had no effect. Irinotecan 59-64 radixin Homo sapiens 17-20 30514181-4 2020 Irinotecan is bioactivated into its metabolite SN-38, which is subsequently detoxified by uridine diphosphate-glucuronosyl transferases (mainly UGT1A1). Irinotecan 47-52 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 144-150 32143347-7 2020 In silico interaction analyses supported the ATPase assay data and suggest that SCO-201 competes with SN-38 for the BCRP drug-binding site. Irinotecan 102-107 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 116-120 32256654-11 2020 In addition, CPT-11 increased the productions of tumor necrosis factor-alpha (TNF-alpha), tumor necrosis factor-beta (TNF-beta), interleukin-6 (IL-6), and interleukin-1 (IL-1), but decreased interleukin-15 (IL-15), interleukin-7 (IL-7), and uridine diphosphate-glucuronosyltransferase 1A1 (UGT1A1). Irinotecan 13-19 tumor necrosis factor Mus musculus 49-76 32256654-11 2020 In addition, CPT-11 increased the productions of tumor necrosis factor-alpha (TNF-alpha), tumor necrosis factor-beta (TNF-beta), interleukin-6 (IL-6), and interleukin-1 (IL-1), but decreased interleukin-15 (IL-15), interleukin-7 (IL-7), and uridine diphosphate-glucuronosyltransferase 1A1 (UGT1A1). Irinotecan 13-19 tumor necrosis factor Mus musculus 78-87 32256654-11 2020 In addition, CPT-11 increased the productions of tumor necrosis factor-alpha (TNF-alpha), tumor necrosis factor-beta (TNF-beta), interleukin-6 (IL-6), and interleukin-1 (IL-1), but decreased interleukin-15 (IL-15), interleukin-7 (IL-7), and uridine diphosphate-glucuronosyltransferase 1A1 (UGT1A1). Irinotecan 13-19 tumor necrosis factor Mus musculus 118-126 32256654-11 2020 In addition, CPT-11 increased the productions of tumor necrosis factor-alpha (TNF-alpha), tumor necrosis factor-beta (TNF-beta), interleukin-6 (IL-6), and interleukin-1 (IL-1), but decreased interleukin-15 (IL-15), interleukin-7 (IL-7), and uridine diphosphate-glucuronosyltransferase 1A1 (UGT1A1). Irinotecan 13-19 interleukin 6 Mus musculus 129-142 32256654-11 2020 In addition, CPT-11 increased the productions of tumor necrosis factor-alpha (TNF-alpha), tumor necrosis factor-beta (TNF-beta), interleukin-6 (IL-6), and interleukin-1 (IL-1), but decreased interleukin-15 (IL-15), interleukin-7 (IL-7), and uridine diphosphate-glucuronosyltransferase 1A1 (UGT1A1). Irinotecan 13-19 interleukin 6 Mus musculus 144-148 32256654-11 2020 In addition, CPT-11 increased the productions of tumor necrosis factor-alpha (TNF-alpha), tumor necrosis factor-beta (TNF-beta), interleukin-6 (IL-6), and interleukin-1 (IL-1), but decreased interleukin-15 (IL-15), interleukin-7 (IL-7), and uridine diphosphate-glucuronosyltransferase 1A1 (UGT1A1). Irinotecan 13-19 interleukin 15 Mus musculus 191-205 32256654-11 2020 In addition, CPT-11 increased the productions of tumor necrosis factor-alpha (TNF-alpha), tumor necrosis factor-beta (TNF-beta), interleukin-6 (IL-6), and interleukin-1 (IL-1), but decreased interleukin-15 (IL-15), interleukin-7 (IL-7), and uridine diphosphate-glucuronosyltransferase 1A1 (UGT1A1). Irinotecan 13-19 interleukin 15 Mus musculus 207-212 32256654-11 2020 In addition, CPT-11 increased the productions of tumor necrosis factor-alpha (TNF-alpha), tumor necrosis factor-beta (TNF-beta), interleukin-6 (IL-6), and interleukin-1 (IL-1), but decreased interleukin-15 (IL-15), interleukin-7 (IL-7), and uridine diphosphate-glucuronosyltransferase 1A1 (UGT1A1). Irinotecan 13-19 interleukin 7 Mus musculus 215-228 32256654-11 2020 In addition, CPT-11 increased the productions of tumor necrosis factor-alpha (TNF-alpha), tumor necrosis factor-beta (TNF-beta), interleukin-6 (IL-6), and interleukin-1 (IL-1), but decreased interleukin-15 (IL-15), interleukin-7 (IL-7), and uridine diphosphate-glucuronosyltransferase 1A1 (UGT1A1). Irinotecan 13-19 interleukin 7 Mus musculus 230-234 32256654-11 2020 In addition, CPT-11 increased the productions of tumor necrosis factor-alpha (TNF-alpha), tumor necrosis factor-beta (TNF-beta), interleukin-6 (IL-6), and interleukin-1 (IL-1), but decreased interleukin-15 (IL-15), interleukin-7 (IL-7), and uridine diphosphate-glucuronosyltransferase 1A1 (UGT1A1). Irinotecan 13-19 UDP glucuronosyltransferase 1 family, polypeptide A1 Mus musculus 241-288 32256654-11 2020 In addition, CPT-11 increased the productions of tumor necrosis factor-alpha (TNF-alpha), tumor necrosis factor-beta (TNF-beta), interleukin-6 (IL-6), and interleukin-1 (IL-1), but decreased interleukin-15 (IL-15), interleukin-7 (IL-7), and uridine diphosphate-glucuronosyltransferase 1A1 (UGT1A1). Irinotecan 13-19 UDP glucuronosyltransferase 1 family, polypeptide A1 Mus musculus 290-296 32170007-1 2020 Irinotecan treats a range of solid tumors, but its effectiveness is severely limited by gastrointestinal (GI) tract toxicity caused by gut bacterial beta-glucuronidase (GUS) enzymes. Irinotecan 0-10 glucuronidase, beta Mus musculus 169-172 32170007-2 2020 Targeted bacterial GUS inhibitors have been shown to partially alleviate irinotecan-induced GI tract damage and resultant diarrhea in mice. Irinotecan 73-83 glucuronidase, beta Mus musculus 19-22 32170007-5 2020 We demonstrate that a single dose of irinotecan increases GI bacterial GUS activity in 1 d and reduces intestinal epithelial cell proliferation in 5 d, both blocked by a single dose of a GUS inhibitor. Irinotecan 37-47 glucuronidase, beta Mus musculus 71-74 32170007-5 2020 We demonstrate that a single dose of irinotecan increases GI bacterial GUS activity in 1 d and reduces intestinal epithelial cell proliferation in 5 d, both blocked by a single dose of a GUS inhibitor. Irinotecan 37-47 glucuronidase, beta Mus musculus 187-190 32170007-6 2020 In a tumor xenograft model, GUS inhibition prevents intestinal toxicity and maintains the antitumor efficacy of irinotecan. Irinotecan 112-122 glucuronidase, beta Mus musculus 28-31 32170007-7 2020 Remarkably, GUS inhibitor also effectively blocks the striking irinotecan-induced bloom of Enterobacteriaceae in immune-deficient mice. Irinotecan 63-73 glucuronidase, beta Mus musculus 12-15 32170007-8 2020 In a genetically engineered mouse model of cancer, GUS inhibition alleviates gut damage, improves survival, and does not alter gut microbial composition; however, by allowing dose intensification, it dramatically improves irinotecan"s effectiveness, reducing tumors to a fraction of that achieved by irinotecan alone, while simultaneously promoting epithelial regeneration. Irinotecan 300-310 glucuronidase, beta Mus musculus 51-54 31857431-0 2020 Phase I Clinical Trial of the Wee1 Inhibitor Adavosertib (AZD1775) with Irinotecan in Children with Relapsed Solid Tumors: A COG Phase I Consortium Report (ADVL1312). Irinotecan 72-82 WEE1 G2 checkpoint kinase Homo sapiens 30-34 31857431-14 2020 CONCLUSIONS: Adavosertib (85 mg/m2) in combination with irinotecan (90 mg/m2) administered orally for 5 days was the MTD in children and adolescents with solid and CNS tumors. Irinotecan 56-66 metallothionein 1E Homo sapiens 117-120 32033066-4 2020 This study analyses the impact of CAPE on the cytotoxic (MTT assay), genotoxic (comet assay) and proapoptotic (caspase-3/7 activity) potential of irinotecan and its metabolite SN-38 in cultures of gastrointestinal neoplastic cells (HCT116, HT29, AGS). Irinotecan 146-156 caspase 3 Homo sapiens 111-122 31769323-11 2020 Progression-free survival on first-line irinotecan chemotherapy was longer in high SATB2 cases (median 8 vs 4 months respectively, p = .019). Irinotecan 40-50 SATB homeobox 2 Homo sapiens 83-88 31189762-5 2020 This linker is more stable in aqueous neutral buffer than a corresponding carbonate-type linker, and releases a payload anti-cancer drug, SN-38, through a two-step sequence upon cathepsin B treatment. Irinotecan 138-143 cathepsin B Homo sapiens 178-189 32269848-7 2020 AGS and NCI-N87 cells transfected with si-RNA-Bcl-3 (si-Bcl-3) showed significantly reduced migratory ability and increased chemosensitivity to oxaliplatin, 5-fluorouracil, and irinotecan. Irinotecan 177-187 BCL3 transcription coactivator Homo sapiens 46-51 31891442-2 2020 The antibody-drug conjugate (ADC) sacituzumab govitecan (SG) targets trophoblast cell-surface antigen-2 (Trop-2) - a cell-surface glycoprotein highly expressed in many epithelial tumors - and delivers the active metabolite of irinotecan SN-38 to Trop-2-positive tumor cells. Irinotecan 226-236 tumor associated calcium signal transducer 2 Homo sapiens 105-111 31891442-2 2020 The antibody-drug conjugate (ADC) sacituzumab govitecan (SG) targets trophoblast cell-surface antigen-2 (Trop-2) - a cell-surface glycoprotein highly expressed in many epithelial tumors - and delivers the active metabolite of irinotecan SN-38 to Trop-2-positive tumor cells. Irinotecan 237-242 tumor associated calcium signal transducer 2 Homo sapiens 105-111 32117765-0 2020 Preclinical Activity of Sacituzumab Govitecan, an Antibody-Drug Conjugate Targeting Trophoblast Cell-Surface Antigen 2 (Trop-2) Linked to the Active Metabolite of Irinotecan (SN-38), in Ovarian Cancer. Irinotecan 163-173 tumor associated calcium signal transducer 2 Homo sapiens 120-126 32117765-0 2020 Preclinical Activity of Sacituzumab Govitecan, an Antibody-Drug Conjugate Targeting Trophoblast Cell-Surface Antigen 2 (Trop-2) Linked to the Active Metabolite of Irinotecan (SN-38), in Ovarian Cancer. Irinotecan 175-180 tumor associated calcium signal transducer 2 Homo sapiens 120-126 32117765-2 2020 Sacituzumab govitecan (SG) is a novel antibody-drug-conjugate (ADC) targeting trophoblast-antigen-2 (Trop-2), a cell surface glycoprotein highly expressed in many epithelial tumors, to deliver SN-38, the active metabolite of irinotecan. Irinotecan 193-198 tumor associated calcium signal transducer 2 Homo sapiens 101-107 32117765-2 2020 Sacituzumab govitecan (SG) is a novel antibody-drug-conjugate (ADC) targeting trophoblast-antigen-2 (Trop-2), a cell surface glycoprotein highly expressed in many epithelial tumors, to deliver SN-38, the active metabolite of irinotecan. Irinotecan 225-235 tumor associated calcium signal transducer 2 Homo sapiens 101-107 32058557-2 2020 Evaluation of pharmacogenomically dosed perioperative gFOLFIRINOX (fluorouracil, leucovorin, oxaliplatin, and UGT1A1 genotype-directed irinotecan) to optimize efficacy while limiting toxic effects may have value. Irinotecan 135-145 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 110-116 32082489-2 2020 Sacituzumab-govitecan (SG) is a new class of antibody-drug-conjugate (ADC) targeting the human-trophoblast-cell-surface marker (Trop-2) conjugated with the active metabolite of irinotecan (SN-38). Irinotecan 177-187 tumor associated calcium signal transducer 2 Homo sapiens 128-134 32082489-2 2020 Sacituzumab-govitecan (SG) is a new class of antibody-drug-conjugate (ADC) targeting the human-trophoblast-cell-surface marker (Trop-2) conjugated with the active metabolite of irinotecan (SN-38). Irinotecan 189-194 tumor associated calcium signal transducer 2 Homo sapiens 128-134 31604667-12 2019 An ARID1A-deficient case that was resistant to multiple cytotoxic drugs, including paclitaxel plus carboplatin in the adjuvant and etoposide plus irinotecan in the first-line treatment, exhibited a dramatic response to gemcitabine in the second-line treatment. Irinotecan 146-156 AT-rich interaction domain 1A Homo sapiens 3-9 31889589-0 2020 Cetuximab enhances the efficiency of irinotecan through simultaneously inhibiting the MAPK signaling and ABCG2 in colorectal cancer cells. Irinotecan 37-47 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 105-110 31889589-3 2020 RESULTS: Cetuximab was found to increase intracellular concentrations of irinotecan as well as cytotoxicity by inhibiting the epidermal growth factor receptor and, by extension, the downstream RAS-RAF-MEK-ERK signaling pathway. Irinotecan 73-83 epidermal growth factor receptor Homo sapiens 126-158 31889589-6 2020 In combination with irinotecan, cetuximab can both significantly induce cell apoptosis by inhibiting the RAS-RAF-MEK-ERK signaling pathway and improve the effects of irinotecan by decreasing drug efflux through the inhibition of ABCG2. Irinotecan 20-30 mitogen-activated protein kinase kinase 7 Homo sapiens 113-116 31889589-6 2020 In combination with irinotecan, cetuximab can both significantly induce cell apoptosis by inhibiting the RAS-RAF-MEK-ERK signaling pathway and improve the effects of irinotecan by decreasing drug efflux through the inhibition of ABCG2. Irinotecan 20-30 mitogen-activated protein kinase 1 Homo sapiens 117-120 31889589-6 2020 In combination with irinotecan, cetuximab can both significantly induce cell apoptosis by inhibiting the RAS-RAF-MEK-ERK signaling pathway and improve the effects of irinotecan by decreasing drug efflux through the inhibition of ABCG2. Irinotecan 20-30 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 229-234 31558477-2 2020 UDP Glucuronosyltransferase 1A1 (UGT1A1) clears SN-38, the active metabolite of irinotecan. Irinotecan 48-53 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-31 31558477-2 2020 UDP Glucuronosyltransferase 1A1 (UGT1A1) clears SN-38, the active metabolite of irinotecan. Irinotecan 48-53 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-39 31558477-2 2020 UDP Glucuronosyltransferase 1A1 (UGT1A1) clears SN-38, the active metabolite of irinotecan. Irinotecan 80-90 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-31 31558477-2 2020 UDP Glucuronosyltransferase 1A1 (UGT1A1) clears SN-38, the active metabolite of irinotecan. Irinotecan 80-90 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-39 31558477-4 2020 We performed a trial to assess the safety and tolerability of FOLFIRABRAX with UGT1A1 genotype-guided dosing of irinotecan. Irinotecan 112-122 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 79-85 31558477-16 2020 CONCLUSIONS: FOLFIRABRAX with genotype-guided dosing of irinotecan is tolerable in patients with advanced gastrointestinal cancer and UGT1A1*1*1 or UGT1A1*1*28 genotypes. Irinotecan 56-66 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 134-140 31558477-16 2020 CONCLUSIONS: FOLFIRABRAX with genotype-guided dosing of irinotecan is tolerable in patients with advanced gastrointestinal cancer and UGT1A1*1*1 or UGT1A1*1*28 genotypes. Irinotecan 56-66 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 148-154 31856090-4 2020 We present the case of a 44-year-old woman with ATM-mutated PC who achieved stable disease as the best response to first-line fluorouracil, leucovorin, irinotecan, and oxaliplatin, followed by progression on a PARP inhibitor. Irinotecan 152-162 ATM serine/threonine kinase Homo sapiens 48-51 31878088-1 2019 Tyrosyl-DNA phosphodiesterase 1 (Tdp1) is an important DNA repair enzyme in humans, and a current and promising inhibition target for the development of new chemosensitizing agents due to its ability to remove DNA damage caused by topoisomerase 1 (Top1) poisons such as topotecan and irinotecan. Irinotecan 284-294 tyrosyl-DNA phosphodiesterase 1 Homo sapiens 0-31 31878088-1 2019 Tyrosyl-DNA phosphodiesterase 1 (Tdp1) is an important DNA repair enzyme in humans, and a current and promising inhibition target for the development of new chemosensitizing agents due to its ability to remove DNA damage caused by topoisomerase 1 (Top1) poisons such as topotecan and irinotecan. Irinotecan 284-294 tyrosyl-DNA phosphodiesterase 1 Homo sapiens 33-37 31878088-1 2019 Tyrosyl-DNA phosphodiesterase 1 (Tdp1) is an important DNA repair enzyme in humans, and a current and promising inhibition target for the development of new chemosensitizing agents due to its ability to remove DNA damage caused by topoisomerase 1 (Top1) poisons such as topotecan and irinotecan. Irinotecan 284-294 DNA topoisomerase I Homo sapiens 231-246 31402291-0 2019 Angiogenic and Antiangiogenic VEGFA Splice Variants in Colorectal Cancer: Prospective Retrospective Cohort Study in Patients Treated With Irinotecan-Based Chemotherapy and Bevacizumab. Irinotecan 138-148 vascular endothelial growth factor A Homo sapiens 30-35 31719636-3 2019 The study aimed to elucidate the effects of insulin (INS), an inexpensive drug with a convincing safety profile, on the susceptibility of colon cancer to chemotherapeutic agents: 5-fluorouracil (FU), oxaliplatin (OXA), irinotecan (IRI), cyclophosphamide (CPA) and docetaxel (DOC). Irinotecan 219-229 insulin Homo sapiens 44-51 31719636-3 2019 The study aimed to elucidate the effects of insulin (INS), an inexpensive drug with a convincing safety profile, on the susceptibility of colon cancer to chemotherapeutic agents: 5-fluorouracil (FU), oxaliplatin (OXA), irinotecan (IRI), cyclophosphamide (CPA) and docetaxel (DOC). Irinotecan 231-234 insulin Homo sapiens 44-51 31704837-8 2019 These results indicated that the simultaneous expression of APEX1 and Jagged-1 might be associated with chemoresistance toward 5-FU, oxaliplatin, and irinotecan. Irinotecan 150-160 apurinic/apyrimidinic endodeoxyribonuclease 1 Homo sapiens 60-65 31704837-8 2019 These results indicated that the simultaneous expression of APEX1 and Jagged-1 might be associated with chemoresistance toward 5-FU, oxaliplatin, and irinotecan. Irinotecan 150-160 jagged canonical Notch ligand 1 Homo sapiens 70-78 31432188-0 2019 The identification of CRNDE, H19, UCA1 and HOTAIR as the key lncRNAs involved in oxaliplatin or irinotecan resistance in the chemotherapy of colorectal cancer based on integrative bioinformatics analysis. Irinotecan 96-106 colorectal neoplasia differentially expressed Homo sapiens 22-27 31598748-0 2019 MiR-218 and miR-100 polymorphisms as markers of irinotecan-based chemotherapy response in metastatic colorectal cancer. Irinotecan 48-58 microRNA 100 Homo sapiens 12-19 31598748-4 2019 METHODS: We investigated the correlation between rs11134527 miR-218 and rs1834306 miR-100 polymorphisms and irinotecan-based regimens with regard to drug efficacy and toxicity. Irinotecan 108-118 microRNA 100 Homo sapiens 82-89 31598748-10 2019 Carriers of the A allele of the miR-218 rs11134527 and T allele of the miR-100 rs1834306 polymorphisms are more likely not to respond to irinotecan-based therapies. Irinotecan 137-147 microRNA 100 Homo sapiens 71-78 31444231-6 2019 Here, we show that loss of LGR5 in colon cancer cells enhanced resistance to irinotecan and 5-fluorouracil and increased expression of adhesion G-protein-coupled receptor, GPR56. Irinotecan 77-87 leucine rich repeat containing G protein-coupled receptor 5 Homo sapiens 27-31 30811654-11 2019 This study found significant changes in the pharmacokinetics of CPT-11 in rats after exposure to X-ray irradiation, and they might be due to significant increases in the expressions of CYP3A1 and CES1. Irinotecan 64-70 cytochrome P450, family 3, subfamily a, polypeptide 23-polypeptide 1 Rattus norvegicus 185-191 30811654-11 2019 This study found significant changes in the pharmacokinetics of CPT-11 in rats after exposure to X-ray irradiation, and they might be due to significant increases in the expressions of CYP3A1 and CES1. Irinotecan 64-70 carboxylesterase 1E Rattus norvegicus 196-200 30612311-0 2019 Sensitization of colorectal cancer to irinotecan therapy by PARP inhibitor rucaparib. Irinotecan 38-48 poly(ADP-ribose) polymerase 1 Homo sapiens 60-64 31432188-0 2019 The identification of CRNDE, H19, UCA1 and HOTAIR as the key lncRNAs involved in oxaliplatin or irinotecan resistance in the chemotherapy of colorectal cancer based on integrative bioinformatics analysis. Irinotecan 96-106 H19 imprinted maternally expressed transcript Homo sapiens 29-32 31432188-0 2019 The identification of CRNDE, H19, UCA1 and HOTAIR as the key lncRNAs involved in oxaliplatin or irinotecan resistance in the chemotherapy of colorectal cancer based on integrative bioinformatics analysis. Irinotecan 96-106 urothelial cancer associated 1 Homo sapiens 34-38 31415918-3 2019 Moreover, medicinal TOP1 inhibitors TPT and SN-38 also augmented IL-10 production significantly, whereas knockdown of TOP1 prevented NC, TPT, and SN-38 from enhancing IL-10 expression. Irinotecan 44-49 interleukin 10 Mus musculus 65-70 31432188-0 2019 The identification of CRNDE, H19, UCA1 and HOTAIR as the key lncRNAs involved in oxaliplatin or irinotecan resistance in the chemotherapy of colorectal cancer based on integrative bioinformatics analysis. Irinotecan 96-106 HOX transcript antisense RNA Homo sapiens 43-49 31222885-5 2019 One example of this type of combination is the addition of the WEE1 inhibitor AZD1775 to the conventional cytotoxic chemotherapeutics, vincristine and irinotecan. Irinotecan 151-161 WEE1 G2 checkpoint kinase Homo sapiens 63-67 31505885-5 2019 The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) cell viability assay found that miR-21 overexpression induced drug resistance to topoisomerase inhibitors (SN-38, doxorubicin, and etoposide/VP-16). Irinotecan 179-184 microRNA 21 Homo sapiens 104-110 31402355-0 2019 Promoting antitumor efficacy by suppressing hypoxia via nano self-assembly of two irinotecan-based dual drug conjugates having a HIF-1alpha inhibitor. Irinotecan 82-92 hypoxia inducible factor 1 subunit alpha Homo sapiens 129-139 31402355-3 2019 To promote the antitumor efficacy of chemical drugs by suppressing hypoxia, we designed and conjugated a hydrophobic HIF-1alpha inhibitor (YC-1) to a hydrophilic anticancer drug, irinotecan (Ir), into one molecular entity via dicarboxylate and PEG3 linkages. Irinotecan 179-189 hypoxia inducible factor 1 subunit alpha Homo sapiens 117-127 31402355-3 2019 To promote the antitumor efficacy of chemical drugs by suppressing hypoxia, we designed and conjugated a hydrophobic HIF-1alpha inhibitor (YC-1) to a hydrophilic anticancer drug, irinotecan (Ir), into one molecular entity via dicarboxylate and PEG3 linkages. Irinotecan 179-189 paternally expressed 3 Homo sapiens 244-248 31415918-3 2019 Moreover, medicinal TOP1 inhibitors TPT and SN-38 also augmented IL-10 production significantly, whereas knockdown of TOP1 prevented NC, TPT, and SN-38 from enhancing IL-10 expression. Irinotecan 146-151 interleukin 10 Mus musculus 167-172 31363167-0 2019 A phase 2 study of panitumumab with irinotecan as salvage therapy in chemorefractory KRAS exon 2 wild-type metastatic colorectal cancer patients. Irinotecan 36-46 KRAS proto-oncogene, GTPase Homo sapiens 85-89 31444410-6 2019 In mechanistic studies, irinotecan not only induces apoptosis by eliciting a DNA damage response, but also acts synergistically with AZD2014 to potentiate the hypophosphorylation of 4E-BP1, a downstream target of mTORC1. Irinotecan 24-34 eukaryotic translation initiation factor 4E binding protein 1 Homo sapiens 182-188 31444410-6 2019 In mechanistic studies, irinotecan not only induces apoptosis by eliciting a DNA damage response, but also acts synergistically with AZD2014 to potentiate the hypophosphorylation of 4E-BP1, a downstream target of mTORC1. Irinotecan 24-34 CREB regulated transcription coactivator 1 Mus musculus 213-219 31466383-9 2019 For example, Sorafenib was applicable for the basal subtype treatment, Irinotecan was optimum for Her2 subtype treatment, Vemurafenib was suitable for the LumA subtype treatment, and Vorinostat could apply to LumB subtype treatment. Irinotecan 71-81 erb-b2 receptor tyrosine kinase 2 Homo sapiens 98-102 31302002-5 2019 Furthermore, IKKalpha or BRAF inhibition synergistically enhances the therapeutic potential of 5-FU and irinotecan to eradicate chemotherapy-resistant metastatic human tumors in vivo. Irinotecan 104-114 component of inhibitor of nuclear factor kappa B kinase complex Homo sapiens 13-21 31302002-5 2019 Furthermore, IKKalpha or BRAF inhibition synergistically enhances the therapeutic potential of 5-FU and irinotecan to eradicate chemotherapy-resistant metastatic human tumors in vivo. Irinotecan 104-114 B-Raf proto-oncogene, serine/threonine kinase Homo sapiens 25-29 31289894-0 2019 Functional mismatch repair and inactive p53 drive sensitization of colorectal cancer cells to irinotecan via the IAP antagonist BV6. Irinotecan 94-104 tumor protein p53 Homo sapiens 40-43 31289894-0 2019 Functional mismatch repair and inactive p53 drive sensitization of colorectal cancer cells to irinotecan via the IAP antagonist BV6. Irinotecan 94-104 alkaline phosphatase, intestinal Homo sapiens 113-116 31289894-2 2019 The purpose of this study was to analyze the cytostatic/cytotoxic response of colorectal carcinoma (CRC) cells to irinotecan, depending on the mismatch repair (MMR) and p53 status and to examine the impact of BV6, a bivalent antagonist of inhibitors of apoptosis c-IAP1/c-IAP2, alone or combined with irinotecan. Irinotecan 114-124 tumor protein p53 Homo sapiens 169-172 31289894-2 2019 The purpose of this study was to analyze the cytostatic/cytotoxic response of colorectal carcinoma (CRC) cells to irinotecan, depending on the mismatch repair (MMR) and p53 status and to examine the impact of BV6, a bivalent antagonist of inhibitors of apoptosis c-IAP1/c-IAP2, alone or combined with irinotecan. Irinotecan 114-124 baculoviral IAP repeat containing 2 Homo sapiens 263-269 31289894-2 2019 The purpose of this study was to analyze the cytostatic/cytotoxic response of colorectal carcinoma (CRC) cells to irinotecan, depending on the mismatch repair (MMR) and p53 status and to examine the impact of BV6, a bivalent antagonist of inhibitors of apoptosis c-IAP1/c-IAP2, alone or combined with irinotecan. Irinotecan 114-124 baculoviral IAP repeat containing 3 Homo sapiens 270-276 31289894-4 2019 Upon irinotecan, MMR-deficient/p53-mutated lines repaired DNA double-strand breaks by homologous recombination less efficiently than MMR-proficient/p53-mutated lines and underwent elevated caspase-9-dependent apoptosis. Irinotecan 5-15 tumor protein p53 Homo sapiens 31-34 31289894-4 2019 Upon irinotecan, MMR-deficient/p53-mutated lines repaired DNA double-strand breaks by homologous recombination less efficiently than MMR-proficient/p53-mutated lines and underwent elevated caspase-9-dependent apoptosis. Irinotecan 5-15 caspase 9 Homo sapiens 189-198 31289894-8 2019 Therefore, the particular MMR+/p53mt signature, often found in non-metastasizing (stage II) CRC might be used as a prognostic factor for an adjuvant therapy using low-dose irinotecan combined with a bivalent IAP antagonist. Irinotecan 172-182 tumor protein p53 Homo sapiens 31-34 31011814-0 2019 SN-38, the active metabolite of irinotecan, inhibits the acute inflammatory response by targeting toll-like receptor 4. Irinotecan 0-5 toll-like receptor 4 Mus musculus 98-118 31011814-0 2019 SN-38, the active metabolite of irinotecan, inhibits the acute inflammatory response by targeting toll-like receptor 4. Irinotecan 32-42 toll-like receptor 4 Mus musculus 98-118 31011814-5 2019 RESULTS: Non-cytotoxic concentrations of SN-38 attenuated LPS (a TLR4 agonist)-driven cell activation without affecting peptidoglycan (a TLR2 agonist)-activating response. Irinotecan 41-46 toll-like receptor 4 Mus musculus 65-69 31011814-8 2019 In addition, SN-38 abrogated LPS-dependent neutrophil migration and reduced TNF-alpha, IL-6, and keratinocyte chemoattractant levels in the air-pouch model, but failed to inhibit zymosan (a TLR2 agonist)-induced cell migration. Irinotecan 13-18 tumor necrosis factor Mus musculus 76-85 31011814-8 2019 In addition, SN-38 abrogated LPS-dependent neutrophil migration and reduced TNF-alpha, IL-6, and keratinocyte chemoattractant levels in the air-pouch model, but failed to inhibit zymosan (a TLR2 agonist)-induced cell migration. Irinotecan 13-18 interleukin 6 Mus musculus 87-91 31011814-8 2019 In addition, SN-38 abrogated LPS-dependent neutrophil migration and reduced TNF-alpha, IL-6, and keratinocyte chemoattractant levels in the air-pouch model, but failed to inhibit zymosan (a TLR2 agonist)-induced cell migration. Irinotecan 13-18 toll-like receptor 2 Mus musculus 190-194 31011814-9 2019 A two-step molecular docking analysis indicated two potential binding sites for the SN-38 in the MD-2/TLR4 complex, the hydrophobic MD-2 pocket (binding energy of - 8.1 kcal/mol) and the rim of the same molecule (- 6.9 kcal/mol). Irinotecan 84-89 toll-like receptor 4 Mus musculus 102-106 31011814-13 2019 CONCLUSIONS: Therefore, SN-38 inhibits acute inflammation by blocking LPS-driven TLR4 signaling. Irinotecan 24-29 toll-like receptor 4 Mus musculus 81-85 31208270-0 2019 Antibody-drug conjugates targeting TROP-2 and incorporating SN-38: A case study of anti-TROP-2 sacituzumab govitecan. Irinotecan 60-65 tumor associated calcium signal transducer 2 Homo sapiens 88-94 31208270-2 2019 SN-38 has been conjugated to a humanized antibody against trophoblast cell surface antigen 2 (TROP-2), which is involved in cancer signaling pathways and has increased expression by many cancer cell types, yielding the ADC sacituzumab govitecan. Irinotecan 0-5 tumor associated calcium signal transducer 2 Homo sapiens 94-100 31141714-0 2019 Comments on: "Clinical utility of ABCB1 genotyping for preventing toxicity in treatment with irinotecan". Irinotecan 93-103 ATP binding cassette subfamily B member 1 Homo sapiens 34-39 31270411-6 2019 The study identified novel off-target interactions by Dactinomycin, Temsirolimus, and Everolimus against NMDA, AMPA, PKA and ERK2, while Irinotecan, Bromocriptine and Dasatinib were top interacting drugs for CaMKII. Irinotecan 137-147 calcium/calmodulin dependent protein kinase II gamma Homo sapiens 208-214 30635626-7 2019 Treatment of mice with AML with irinotecan, known to inhibit topoisomerase I and FUBP1, significantly prolonged survival alone or in combination with cytarabine. Irinotecan 32-42 far upstream element (FUSE) binding protein 1 Mus musculus 81-86 30831128-0 2019 Intestinal bacterial beta-glucuronidase as a possible predictive biomarker of irinotecan-induced diarrhea severity. Irinotecan 78-88 glucuronidase beta Homo sapiens 21-39 30831128-4 2019 Recent studies have highlighted the important role of the intestinal bacterial beta-glucuronidase (BGUS) in the onset of irinotecan-induced diarrhea. Irinotecan 121-131 glucuronidase beta Homo sapiens 79-97 30831128-4 2019 Recent studies have highlighted the important role of the intestinal bacterial beta-glucuronidase (BGUS) in the onset of irinotecan-induced diarrhea. Irinotecan 121-131 glucuronidase beta Homo sapiens 99-103 30831128-6 2019 BGUS selective inhibitors that are currently in development may alleviate irinotecan-induced diarrhea, and may help to reduce its morbidity and enhance its activity. Irinotecan 74-84 glucuronidase beta Homo sapiens 0-4 30831128-8 2019 In addition, we hypothesize that using BGUS activity as a predictive biomarker of irinotecan-induced diarrhea severity will help to select cancer patients for treatment with irinotecan chemotherapy (whether at full or adapted dose). Irinotecan 82-92 glucuronidase beta Homo sapiens 39-43 30831128-8 2019 In addition, we hypothesize that using BGUS activity as a predictive biomarker of irinotecan-induced diarrhea severity will help to select cancer patients for treatment with irinotecan chemotherapy (whether at full or adapted dose). Irinotecan 174-184 glucuronidase beta Homo sapiens 39-43 30603911-0 2019 Association between UGT1A1 gene polymorphism and safety and efficacy of irinotecan monotherapy as the third-line treatment for advanced gastric cancer. Irinotecan 72-82 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 20-26 30603911-1 2019 BACKGROUND: While uridine diphosphate glucuronosyltransferase (UGT) 1A1 is a key enzyme in the metabolism of irinotecan, relationship between UGT1A1 genotype and safety and efficacy of irinotecan monotherapy in patients with advanced gastric cancer is not clarified. Irinotecan 109-119 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 18-71 30603911-1 2019 BACKGROUND: While uridine diphosphate glucuronosyltransferase (UGT) 1A1 is a key enzyme in the metabolism of irinotecan, relationship between UGT1A1 genotype and safety and efficacy of irinotecan monotherapy in patients with advanced gastric cancer is not clarified. Irinotecan 185-195 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 142-148 30603911-8 2019 CONCLUSIONS: The UGT1A1 polymorphism may be related to the clinical outcomes of irinotecan monotherapy as the third-line treatment for advanced gastric cancer. Irinotecan 80-90 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 17-23 31141715-0 2019 Comments on: "Clinical utility of ABCB1 genotyping for preventing toxicity in treatment with irinotecan". Irinotecan 93-103 ATP binding cassette subfamily B member 1 Homo sapiens 34-39 31217818-0 2019 An expansion study of genotype-driven weekly irinotecan and capecitabine in combination with neoadjuvant radiotherapy for locally advanced rectal cancer with UGT1A1 *1*1 genotype. Irinotecan 45-55 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 158-164 31243264-6 2019 We found that the interaction between GRP78 and SPARC increased during exposure to 5-FU, CPT-11, and tunicamycin, resulting in an attenuation of GRP78"s inhibition of apoptosis. Irinotecan 89-95 heat shock protein family A (Hsp70) member 5 Homo sapiens 38-43 31243264-6 2019 We found that the interaction between GRP78 and SPARC increased during exposure to 5-FU, CPT-11, and tunicamycin, resulting in an attenuation of GRP78"s inhibition of apoptosis. Irinotecan 89-95 secreted protein acidic and cysteine rich Homo sapiens 48-53 31243264-6 2019 We found that the interaction between GRP78 and SPARC increased during exposure to 5-FU, CPT-11, and tunicamycin, resulting in an attenuation of GRP78"s inhibition of apoptosis. Irinotecan 89-95 heat shock protein family A (Hsp70) member 5 Homo sapiens 145-150 31056264-0 2019 A novel mechanism of irinotecan targeting MDM2 and Bcl-xL. Irinotecan 21-31 MDM2 proto-oncogene Homo sapiens 42-46 31056264-0 2019 A novel mechanism of irinotecan targeting MDM2 and Bcl-xL. Irinotecan 21-31 BCL2 like 1 Homo sapiens 51-57 31056264-2 2019 In this study, we present a new mechanism of Irinotecan which inhibits the activities of MDM2, an E3 ligase of tumour suppressor p53, and Bcl-xL, an anti-apoptotic protein, through direct binding. Irinotecan 45-55 MDM2 proto-oncogene Homo sapiens 89-93 31056264-2 2019 In this study, we present a new mechanism of Irinotecan which inhibits the activities of MDM2, an E3 ligase of tumour suppressor p53, and Bcl-xL, an anti-apoptotic protein, through direct binding. Irinotecan 45-55 tumor protein p53 Homo sapiens 129-132 31056264-2 2019 In this study, we present a new mechanism of Irinotecan which inhibits the activities of MDM2, an E3 ligase of tumour suppressor p53, and Bcl-xL, an anti-apoptotic protein, through direct binding. Irinotecan 45-55 BCL2 like 1 Homo sapiens 138-144 31056264-3 2019 In our structure modelling study, Irinotecan could fit to the binding sites of MDM2 and Bcl-xL for their known drugs, Nutlin-3 and ABT-737, with a better binding affinity than to Topo I. Irinotecan 34-44 MDM2 proto-oncogene Homo sapiens 79-83 31056264-3 2019 In our structure modelling study, Irinotecan could fit to the binding sites of MDM2 and Bcl-xL for their known drugs, Nutlin-3 and ABT-737, with a better binding affinity than to Topo I. Irinotecan 34-44 BCL2 like 1 Homo sapiens 88-94 31056264-5 2019 We further showed that Irinotecan increased the amount of p53 only in the presence of MDM2 and inhibited the physical interaction of Bcl-xL with Bim, a core pro-apoptotic protein. Irinotecan 23-33 tumor protein p53 Homo sapiens 58-61 31056264-5 2019 We further showed that Irinotecan increased the amount of p53 only in the presence of MDM2 and inhibited the physical interaction of Bcl-xL with Bim, a core pro-apoptotic protein. Irinotecan 23-33 MDM2 proto-oncogene Homo sapiens 86-90 31056264-5 2019 We further showed that Irinotecan increased the amount of p53 only in the presence of MDM2 and inhibited the physical interaction of Bcl-xL with Bim, a core pro-apoptotic protein. Irinotecan 23-33 BCL2 like 1 Homo sapiens 133-139 31056264-5 2019 We further showed that Irinotecan increased the amount of p53 only in the presence of MDM2 and inhibited the physical interaction of Bcl-xL with Bim, a core pro-apoptotic protein. Irinotecan 23-33 BCL2 like 11 Homo sapiens 145-148 31056264-7 2019 Collectively, we suggest a new mechanism of action for Irinotecan as a dual target inhibitor of MDM2 and Bcl-xL facilitating the anticancer activities mediated by p53 and Bcl-xL interaction partners. Irinotecan 55-65 MDM2 proto-oncogene Homo sapiens 96-100 31056264-7 2019 Collectively, we suggest a new mechanism of action for Irinotecan as a dual target inhibitor of MDM2 and Bcl-xL facilitating the anticancer activities mediated by p53 and Bcl-xL interaction partners. Irinotecan 55-65 BCL2 like 1 Homo sapiens 105-111 31056264-7 2019 Collectively, we suggest a new mechanism of action for Irinotecan as a dual target inhibitor of MDM2 and Bcl-xL facilitating the anticancer activities mediated by p53 and Bcl-xL interaction partners. Irinotecan 55-65 tumor protein p53 Homo sapiens 163-166 31056264-7 2019 Collectively, we suggest a new mechanism of action for Irinotecan as a dual target inhibitor of MDM2 and Bcl-xL facilitating the anticancer activities mediated by p53 and Bcl-xL interaction partners. Irinotecan 55-65 BCL2 like 1 Homo sapiens 171-177 31242600-2 2019 As torsional stress is generated during transcription by progression of RNA polymerase II through the transcribed gene, we tested the effects of camptothecin and of the approved TOP1 inhibitors Topotecan and SN-38 on TNFalpha-induced gene expression. Irinotecan 208-213 tumor necrosis factor Homo sapiens 217-225 31217818-1 2019 Background: In our previous dose-escalation study, we uncovered the maximum tolerated dose (MTD) of weekly irinotecan was escalated to 80 mg/m2 and 65 mg/m2 for UDP glucuronosyltransferase family 1 member A1 (UGT1A1) *1*1 and *1*28 rectal cancer patients in neoadjuvant chemoradiotherapy (nCRT). Irinotecan 107-117 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 161-207 31217818-1 2019 Background: In our previous dose-escalation study, we uncovered the maximum tolerated dose (MTD) of weekly irinotecan was escalated to 80 mg/m2 and 65 mg/m2 for UDP glucuronosyltransferase family 1 member A1 (UGT1A1) *1*1 and *1*28 rectal cancer patients in neoadjuvant chemoradiotherapy (nCRT). Irinotecan 107-117 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 209-215 30953637-8 2019 Moreover, the combination of SN-38 and sunitinib caused synergism on colon cancer cells, with significant inhibition of the ABCG2 gene expression and an increase of SN-38 intracellular concentrations. Irinotecan 29-34 ATP binding cassette subfamily G member 2 (Junior blood group) Mus musculus 124-129 30968312-9 2019 For illustration purpose, the approaches were also applied to an irinotecan mixture model demonstrating 36% lower clearance of irinotecan metabolite (SN-38) in individuals with UGT1A1 homo/heterozygote versus wild-type genotype. Irinotecan 65-75 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 177-183 31011934-0 2019 Cytotoxic synergy between alisertib and carboplatin versus alisertib and irinotecan are inversely dependent on MGMT levels in glioblastoma cells. Irinotecan 73-83 O-6-methylguanine-DNA methyltransferase Homo sapiens 111-115 31011934-7 2019 MGMT knockdown increased apoptosis caused by combined alisertib and irinotecan, while exogenous MGMT overexpression increased apoptosis from alisertib and carboplatin combination treatment. Irinotecan 68-78 O-6-methylguanine-DNA methyltransferase Homo sapiens 0-4 30968312-9 2019 For illustration purpose, the approaches were also applied to an irinotecan mixture model demonstrating 36% lower clearance of irinotecan metabolite (SN-38) in individuals with UGT1A1 homo/heterozygote versus wild-type genotype. Irinotecan 127-137 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 177-183 30968312-9 2019 For illustration purpose, the approaches were also applied to an irinotecan mixture model demonstrating 36% lower clearance of irinotecan metabolite (SN-38) in individuals with UGT1A1 homo/heterozygote versus wild-type genotype. Irinotecan 150-155 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 177-183 31126331-13 2019 CONCLUSIONS: Treatment with CMAB009 plus irinotecan was found to be a superior second-line regimen in comparison to irinotecan-only in KRAS wild-type mCRC patients. Irinotecan 41-51 KRAS proto-oncogene, GTPase Homo sapiens 135-139 31067275-5 2019 We now further demonstrate that high levels of HMGA2 also protected cancer cells against DNA breaks triggered by the clinically important TOP1 poison irinotecan. Irinotecan 150-160 high mobility group AT-hook 2 Homo sapiens 47-52 30645764-2 2019 UGT1A1 (UDP glucuronosyltransferase 1A1) eliminates the active metabolite of irinotecan. Irinotecan 77-87 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 30645764-2 2019 UGT1A1 (UDP glucuronosyltransferase 1A1) eliminates the active metabolite of irinotecan. Irinotecan 77-87 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 8-39 30645764-6 2019 Irinotecan doses of 180, 135, and 90 mg/m2 were administered for UGT1A1 genotypes *1/*1, *1/*28, and *28/*28, respectively. Irinotecan 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 65-71 30925062-5 2019 In addition, the thienopyrimidine could also sensitize ABCB1- as well as ABCG2-overexpressing cells toward daunorubicin and SN-38, respectively, in concentration ranges that qualified it as one of the ten best triple ABCC1/ABCB1/ABCG2 inhibitors in the literature. Irinotecan 124-129 ATP binding cassette subfamily B member 1 Homo sapiens 55-60 30925062-5 2019 In addition, the thienopyrimidine could also sensitize ABCB1- as well as ABCG2-overexpressing cells toward daunorubicin and SN-38, respectively, in concentration ranges that qualified it as one of the ten best triple ABCC1/ABCB1/ABCG2 inhibitors in the literature. Irinotecan 124-129 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 73-78 30925062-5 2019 In addition, the thienopyrimidine could also sensitize ABCB1- as well as ABCG2-overexpressing cells toward daunorubicin and SN-38, respectively, in concentration ranges that qualified it as one of the ten best triple ABCC1/ABCB1/ABCG2 inhibitors in the literature. Irinotecan 124-129 ATP binding cassette subfamily C member 1 Homo sapiens 217-222 30925062-5 2019 In addition, the thienopyrimidine could also sensitize ABCB1- as well as ABCG2-overexpressing cells toward daunorubicin and SN-38, respectively, in concentration ranges that qualified it as one of the ten best triple ABCC1/ABCB1/ABCG2 inhibitors in the literature. Irinotecan 124-129 ATP binding cassette subfamily B member 1 Homo sapiens 223-228 30925062-5 2019 In addition, the thienopyrimidine could also sensitize ABCB1- as well as ABCG2-overexpressing cells toward daunorubicin and SN-38, respectively, in concentration ranges that qualified it as one of the ten best triple ABCC1/ABCB1/ABCG2 inhibitors in the literature. Irinotecan 124-129 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 229-234 31067275-7 2019 In contrast, low to moderate HMGA2 protein levels surprisingly potentiated the formation of irinotecan-induced genotoxic covalent TOP1-DNA cleavage complexes. Irinotecan 92-102 high mobility group AT-hook 2 Homo sapiens 29-34 31067275-10 2019 These findings were corroborated by an increased irinotecan sensitivity of patient-derived xenografts of colorectal cancers exhibiting low to moderate HMGA2 levels. Irinotecan 49-59 high mobility group AT-hook 2 Homo sapiens 151-156 30810774-6 2019 RESULTS: Concentration-dependent inhibition of SN-38 glucuronidation was observed in the HLMs and recombinant human UGT1A1 experiments: Pazopanib noncompetitively inhibited SN-38 glucuronidation by HLMs (Ki,u = 1.6 +- 0.05 microM) and recombinant human UGT1A1 (Ki,u = 0.69 +- 0.02 microM). Irinotecan 47-52 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 116-122 30898612-6 2019 Interestingly, co-treatment with nutlin-3a and certain chemotherapeutic drug such as irinotecan and oxaliplatin resulted in antagonistic effects in cells both lacking and containing WT-TP53 activity. Irinotecan 85-95 tumor protein p53 Homo sapiens 185-189 30840064-3 2019 PATIENTS AND METHODS: This was a single-arm phase II trial that analysed 38 KRAS, NRAS, BRAF and PIK3CA hotspots in tumour tissues of irinotecan-resistant metastatic colorectal cancer patients who received panitumumab plus FOLFIRI until disease progression or early withdrawal. Irinotecan 134-144 KRAS proto-oncogene, GTPase Homo sapiens 76-80 30840064-3 2019 PATIENTS AND METHODS: This was a single-arm phase II trial that analysed 38 KRAS, NRAS, BRAF and PIK3CA hotspots in tumour tissues of irinotecan-resistant metastatic colorectal cancer patients who received panitumumab plus FOLFIRI until disease progression or early withdrawal. Irinotecan 134-144 NRAS proto-oncogene, GTPase Homo sapiens 82-86 30840064-3 2019 PATIENTS AND METHODS: This was a single-arm phase II trial that analysed 38 KRAS, NRAS, BRAF and PIK3CA hotspots in tumour tissues of irinotecan-resistant metastatic colorectal cancer patients who received panitumumab plus FOLFIRI until disease progression or early withdrawal. Irinotecan 134-144 B-Raf proto-oncogene, serine/threonine kinase Homo sapiens 88-92 30840064-3 2019 PATIENTS AND METHODS: This was a single-arm phase II trial that analysed 38 KRAS, NRAS, BRAF and PIK3CA hotspots in tumour tissues of irinotecan-resistant metastatic colorectal cancer patients who received panitumumab plus FOLFIRI until disease progression or early withdrawal. Irinotecan 134-144 phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha Homo sapiens 97-103 30810774-0 2019 Pazopanib interacts with irinotecan by inhibiting UGT1A1-mediated glucuronidation, but not OATP1B1-mediated hepatic uptake, of an active metabolite SN-38. Irinotecan 25-35 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 50-56 30810774-6 2019 RESULTS: Concentration-dependent inhibition of SN-38 glucuronidation was observed in the HLMs and recombinant human UGT1A1 experiments: Pazopanib noncompetitively inhibited SN-38 glucuronidation by HLMs (Ki,u = 1.6 +- 0.05 microM) and recombinant human UGT1A1 (Ki,u = 0.69 +- 0.02 microM). Irinotecan 47-52 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 253-259 30810774-10 2019 CONCLUSIONS: Results showed that pazopanib inhibits UGT1A1-mediated SN-38 glucuronidation, but not OATP1B1-mediated SN-38 uptake. Irinotecan 68-73 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 52-58 30815720-0 2019 Irinotecan induces enterocyte cell death and changes to muc2 and muc4 composition during mucositis in a tumour-bearing DA rat model. Irinotecan 0-10 mucin 2, oligomeric mucus/gel-forming Rattus norvegicus 56-60 30815720-0 2019 Irinotecan induces enterocyte cell death and changes to muc2 and muc4 composition during mucositis in a tumour-bearing DA rat model. Irinotecan 0-10 mucin 4, cell surface associated Rattus norvegicus 65-69 30782852-2 2019 In the past decade, OATPs, primarily OATP1A2, OATP1B1, and OATP1B3, have emerged as potential mediators of chemotherapy disposition, including drugs such as methotrexate, doxorubicin, paclitaxel, docetaxel, irinotecan and its important metabolite 7-ethyl-10-hydroxycamptothecin, and certain tyrosine kinase inhibitors. Irinotecan 207-217 solute carrier organic anion transporter family member 1A2 Homo sapiens 37-44 30782852-2 2019 In the past decade, OATPs, primarily OATP1A2, OATP1B1, and OATP1B3, have emerged as potential mediators of chemotherapy disposition, including drugs such as methotrexate, doxorubicin, paclitaxel, docetaxel, irinotecan and its important metabolite 7-ethyl-10-hydroxycamptothecin, and certain tyrosine kinase inhibitors. Irinotecan 207-217 solute carrier organic anion transporter family member 1B3 Homo sapiens 59-66 30782852-2 2019 In the past decade, OATPs, primarily OATP1A2, OATP1B1, and OATP1B3, have emerged as potential mediators of chemotherapy disposition, including drugs such as methotrexate, doxorubicin, paclitaxel, docetaxel, irinotecan and its important metabolite 7-ethyl-10-hydroxycamptothecin, and certain tyrosine kinase inhibitors. Irinotecan 247-277 solute carrier organic anion transporter family member 1A2 Homo sapiens 37-44 30782852-2 2019 In the past decade, OATPs, primarily OATP1A2, OATP1B1, and OATP1B3, have emerged as potential mediators of chemotherapy disposition, including drugs such as methotrexate, doxorubicin, paclitaxel, docetaxel, irinotecan and its important metabolite 7-ethyl-10-hydroxycamptothecin, and certain tyrosine kinase inhibitors. Irinotecan 247-277 solute carrier organic anion transporter family member 1B3 Homo sapiens 59-66 32259055-6 2019 S phase arrest was induced with the topoisomerase I inhibitor SN38; the addition of CHK1i rapidly activated CDK2, inducing S phase progression that was inhibited by the CDK2 inhibitor CVT-313. Irinotecan 62-66 checkpoint kinase 1 Homo sapiens 84-89 31105879-3 2019 Acute and chronic exposition of colon cancer cell lines to CT/CTE PEDF-derived peptides decreased drug-resistance to conventional colorectal cancer treatments, such as oxaliplatin or irinotecan. Irinotecan 183-193 serpin family F member 1 Homo sapiens 66-70 32259055-6 2019 S phase arrest was induced with the topoisomerase I inhibitor SN38; the addition of CHK1i rapidly activated CDK2, inducing S phase progression that was inhibited by the CDK2 inhibitor CVT-313. Irinotecan 62-66 cyclin dependent kinase 2 Homo sapiens 169-173 30968152-1 2019 Irinotecan (CPT-11) is a DNA topoisomerase I inhibitor which is widely used in clinical chemotherapy, particularly for colorectal cancer treatment. Irinotecan 0-10 DNA topoisomerase I Rattus norvegicus 25-44 31164525-2 2019 Since both patients were revealed to harbor UGT1A1 polymorphisms, which were highly associated with irinotecan-induced toxicity(the former: UGT1A1 *6/*28, the latter: UGT1A1*6/*6), there was no alternative hopeful treatment other than FOLFIRINOX for them. Irinotecan 100-110 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 44-50 31164525-2 2019 Since both patients were revealed to harbor UGT1A1 polymorphisms, which were highly associated with irinotecan-induced toxicity(the former: UGT1A1 *6/*28, the latter: UGT1A1*6/*6), there was no alternative hopeful treatment other than FOLFIRINOX for them. Irinotecan 100-110 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 140-146 31164525-2 2019 Since both patients were revealed to harbor UGT1A1 polymorphisms, which were highly associated with irinotecan-induced toxicity(the former: UGT1A1 *6/*28, the latter: UGT1A1*6/*6), there was no alternative hopeful treatment other than FOLFIRINOX for them. Irinotecan 100-110 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 140-146 30952738-0 2019 Cytoplasmic E-Cadherin Expression Is Associated With Higher Tumour Level of VEGFA, Lower Response Rate to Irinotecan-based Treatment and Poorer Prognosis in Patients With Metastatic Colorectal Cancer. Irinotecan 106-116 cadherin 1 Homo sapiens 12-22 30952738-10 2019 In addition, among the patients with intense VEGFA expression (n=36), those who had positive cytoplasmic E-cadherin in their tumours had a lower response-rate to first-line therapy with irinotecan, fluorouracil and leucovorin regimen: 5 out of 36 (14%) were chemosensitive. Irinotecan 186-196 cadherin 1 Homo sapiens 105-115 30339275-7 2019 In patients receiving fluoropyrimidine/irinotecan, the incremental cost between DPYD variant and UGT1A1*28/*28 carriers and noncarriers was $2,975. Irinotecan 39-49 dihydropyrimidine dehydrogenase Homo sapiens 80-84 30339275-7 2019 In patients receiving fluoropyrimidine/irinotecan, the incremental cost between DPYD variant and UGT1A1*28/*28 carriers and noncarriers was $2,975. Irinotecan 39-49 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 97-103 30968152-1 2019 Irinotecan (CPT-11) is a DNA topoisomerase I inhibitor which is widely used in clinical chemotherapy, particularly for colorectal cancer treatment. Irinotecan 12-18 DNA topoisomerase I Rattus norvegicus 25-44 30415007-0 2019 Combined treatment of ABT199 and irinotecan suppresses KRAS-mutant lung cancer cells. Irinotecan 33-43 KRAS proto-oncogene, GTPase Homo sapiens 55-59 30881507-2 2019 Expression of CYP3A5, which is involved in the degradation of irinotecan, has also been reported in colorectal cancer (CRC). Irinotecan 62-72 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 14-20 30881507-10 2019 A statistically significant inverse correlation was identified between CYP3A5 expression in CRC tissues and tumor response to irinotecan therapy. Irinotecan 126-136 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 71-77 30881507-12 2019 As CYP3A5 is involved in the degradation of irinotecan, the significantly higher intratumoral expression of CYP3A5 in patients with CRC who do not respond to irinotecan-based chemotherapy may indicate a causal role of CYP3A5 in tumor resistance. Irinotecan 44-54 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 3-9 30881507-12 2019 As CYP3A5 is involved in the degradation of irinotecan, the significantly higher intratumoral expression of CYP3A5 in patients with CRC who do not respond to irinotecan-based chemotherapy may indicate a causal role of CYP3A5 in tumor resistance. Irinotecan 158-168 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 108-114 30881507-12 2019 As CYP3A5 is involved in the degradation of irinotecan, the significantly higher intratumoral expression of CYP3A5 in patients with CRC who do not respond to irinotecan-based chemotherapy may indicate a causal role of CYP3A5 in tumor resistance. Irinotecan 158-168 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 108-114 30881507-0 2019 Tumor response to irinotecan is associated with CYP3A5 expression in colorectal cancer. Irinotecan 18-28 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 48-54 30476968-16 2019 Conclusions and Relevance: This is the first prospective demonstration that a rechallenge strategy with cetuximab and irinotecan may be active in patients with RAS and BRAF wild-type mCRC with acquired resistance to first-line irinotecan- and cetuximab-based therapy. Irinotecan 118-128 B-Raf proto-oncogene, serine/threonine kinase Homo sapiens 168-172 29884612-10 2019 Finally, EAC cell lines showed subtype-specific responses to topotecan, SN-38 and palbociclib treatment. Irinotecan 72-77 CYLD lysine 63 deubiquitinase Homo sapiens 9-12 30602616-4 2019 We present the case of a recurrent nasoethmoidal ITAC informed as RAS and BRAF wild-type by standard real-time polymerase chain reaction methods and treated with first-line cetuximab and irinotecan without response. Irinotecan 187-197 C-X-C motif chemokine ligand 11 Homo sapiens 49-53 30602616-4 2019 We present the case of a recurrent nasoethmoidal ITAC informed as RAS and BRAF wild-type by standard real-time polymerase chain reaction methods and treated with first-line cetuximab and irinotecan without response. Irinotecan 187-197 B-Raf proto-oncogene, serine/threonine kinase Homo sapiens 74-78 30820689-8 2019 In pharmacokinetics studies, for pro-drug IRT, the t1/2beta of IRT-PLGA-NPs was extended from 0.483 to 3.327 h compared with irinotecan solution (IRT-Sol), and for its active metabolite SN-38, the t1/2beta was extended from 1.889 to 4.811 h, which indicated that IRT-PLGA-NPs could prolong the retention times of both IRT and SN-38. Irinotecan 186-191 interleukin 1 receptor like 1 Homo sapiens 51-59 30899442-7 2019 In vitro CD133 expression and its co-expression with CD44 were associated with primary-resistance to irinotecan and acquired-resistance to anti-EGFR inhibitors respectively. Irinotecan 101-111 prominin 1 Homo sapiens 9-14 30899442-7 2019 In vitro CD133 expression and its co-expression with CD44 were associated with primary-resistance to irinotecan and acquired-resistance to anti-EGFR inhibitors respectively. Irinotecan 101-111 CD44 molecule (Indian blood group) Homo sapiens 53-57 30820689-8 2019 In pharmacokinetics studies, for pro-drug IRT, the t1/2beta of IRT-PLGA-NPs was extended from 0.483 to 3.327 h compared with irinotecan solution (IRT-Sol), and for its active metabolite SN-38, the t1/2beta was extended from 1.889 to 4.811 h, which indicated that IRT-PLGA-NPs could prolong the retention times of both IRT and SN-38. Irinotecan 326-331 interleukin 1 receptor like 1 Homo sapiens 51-59 30315927-0 2019 A molecular basis for the synergy between 17-allylamino-17-demethoxy geldanamycin with Capecitabine and Irinotecan in human colorectal cancer cells through VEFG and MMP-9 gene expression. Irinotecan 104-114 matrix metallopeptidase 9 Homo sapiens 165-170 30447099-1 2019 Studies have indicated an association between UDP-glucuronosyltransferase-1A1 (UGT1A1) genetic polymorphisms and irinotecan-induced toxicity. Irinotecan 113-123 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 79-85 30447099-2 2019 We undertook this study to investigate the association between UGT1A1 genetic polymorphisms and toxicity in patients treated with the FOLFIRINOX (comprising oxaliplatin, irinotecan, fluorouracil, and leucovorin) chemotherapy regimen in the JASPAC 06 study. Irinotecan 170-180 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 63-69 30447099-1 2019 Studies have indicated an association between UDP-glucuronosyltransferase-1A1 (UGT1A1) genetic polymorphisms and irinotecan-induced toxicity. Irinotecan 113-123 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 46-77 30696915-8 2019 Knockdown of Crabp2 further inhibited the growth of cancer cells as compared with that by gemcitabine or irinotecan alone. Irinotecan 105-115 cellular retinoic acid binding protein 2 Homo sapiens 13-19 30733972-0 2019 A Novel Anti-EGFR mAb Ame55 with Lower Toxicity and Better Efficacy than Cetuximab When Combined with Irinotecan. Irinotecan 102-112 epidermal growth factor receptor Homo sapiens 13-17 30634695-7 2019 These results indicate that the substitution of Gly at position 187 of ABCC4 to Trp resulted in reduced SN-38 resistance. Irinotecan 104-109 ATP binding cassette subfamily C member 4 Homo sapiens 71-76 30551482-11 2019 Additionally, a potent inhibitory effect of GQT extract against hCE2 was observedin vitro, with its IC50 value of 0.187 mg/ml, suggesting alleviating activity on hCE2-mediated severe diarrhea in patients suffered from CPT-11. Irinotecan 218-224 carboxylesterase 2 Homo sapiens 64-68 30662270-0 2019 Complete response with fluorouracil and irinotecan with a BRAFV600E and EGFR inhibitor in BRAF-mutated metastatic colorectal cancer: a case report. Irinotecan 40-50 epidermal growth factor receptor Homo sapiens 72-76 30662270-0 2019 Complete response with fluorouracil and irinotecan with a BRAFV600E and EGFR inhibitor in BRAF-mutated metastatic colorectal cancer: a case report. Irinotecan 40-50 B-Raf proto-oncogene, serine/threonine kinase Homo sapiens 58-62 30662270-9 2019 Conclusion: The combination of fluorouracil and irinotecan with a BRAFV600E and EGFR inhibitor may have synergistic action, leading to recession of secondary metastases in patients with BRAFV600E-mutated colorectal cancer. Irinotecan 48-58 epidermal growth factor receptor Homo sapiens 80-84 30551482-11 2019 Additionally, a potent inhibitory effect of GQT extract against hCE2 was observedin vitro, with its IC50 value of 0.187 mg/ml, suggesting alleviating activity on hCE2-mediated severe diarrhea in patients suffered from CPT-11. Irinotecan 218-224 carboxylesterase 2 Homo sapiens 162-166 30377777-11 2019 CONCLUSION: The present study demonstrated that dose reduction by 20% ensured safety and efficacy of irinotecan in mCRC patients with homozygous mutation in UGT1A1 genes. Irinotecan 101-111 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 157-163 30585257-1 2019 BACKGROUND: Patients harbouring the UGT1A1*28/*28 genotype are at risk of severe toxicity with the standard irinotecan dose. Irinotecan 108-118 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 36-42 30585257-6 2019 In the experimental group, the irinotecan dose was 300 mg/m2 for UGT1A1*1/*1 and 260 mg/m2 for *1/*28 patients. Irinotecan 31-41 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 65-71 30585257-13 2019 CONCLUSIONS: Patients with the UGT1A1*1/*1 and *1/*28 genotypes can receive high doses of irinotecan to achieve a more favourable ORR without significant adverse events. Irinotecan 90-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 31-37 30307354-2 2019 CASE PRESENTATION: Here we present a KRAS/NRAS/BRAF wild-type mCRC patient who has been previously treated with FOLFIRI (fluorouracil, leucovorin, and irinotecan), XELOX (capecitabine and oxaliplatin), cetuximab and bevacizumab, and then received the next generation sequencing (NGS) and whose metastatic subcutaneous nodule was resected to generate patient-derived xenograft (PDX) models. Irinotecan 151-161 KRAS proto-oncogene, GTPase Homo sapiens 37-41 30377777-0 2019 Dose adjustment of irinotecan based on UGT1A1 polymorphisms in patients with colorectal cancer. Irinotecan 19-29 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 39-45 31045514-4 2019 OBJECTIVE: Investigation of the association between rs4759314 HOTAIR and rs3200401 MALAT1 polymorphisms and irinotecan-based chemotherapy in terms of drug efficacy and toxicity. Irinotecan 108-118 HOX transcript antisense RNA Homo sapiens 62-68 30377777-2 2019 SN-38 is an active metabolite of irinotecan, which is formed by carboxylesterase and inactivated by UDP-glucuronyltransferase (UGT) 1A1. Irinotecan 0-5 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 100-135 30377777-2 2019 SN-38 is an active metabolite of irinotecan, which is formed by carboxylesterase and inactivated by UDP-glucuronyltransferase (UGT) 1A1. Irinotecan 33-43 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 100-135 30377777-3 2019 The UGT enzyme activity is reduced in patients with homozygous mutation in UGT1A1 genes (*6/*6, *28/*28 and *6/*28); thus dose reduction is required for prevention of severe adverse events associated with irinotecan. Irinotecan 205-215 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 4-7 30377777-3 2019 The UGT enzyme activity is reduced in patients with homozygous mutation in UGT1A1 genes (*6/*6, *28/*28 and *6/*28); thus dose reduction is required for prevention of severe adverse events associated with irinotecan. Irinotecan 205-215 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 75-81 30377777-4 2019 The present study was designed to investigate the relationship between UGT1A1 polymorphisms and the incidence of adverse events or the therapeutic effect in mCRC patients who received irinotecan. Irinotecan 184-194 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 71-77 30307354-2 2019 CASE PRESENTATION: Here we present a KRAS/NRAS/BRAF wild-type mCRC patient who has been previously treated with FOLFIRI (fluorouracil, leucovorin, and irinotecan), XELOX (capecitabine and oxaliplatin), cetuximab and bevacizumab, and then received the next generation sequencing (NGS) and whose metastatic subcutaneous nodule was resected to generate patient-derived xenograft (PDX) models. Irinotecan 151-161 NRAS proto-oncogene, GTPase Homo sapiens 42-46 30307354-2 2019 CASE PRESENTATION: Here we present a KRAS/NRAS/BRAF wild-type mCRC patient who has been previously treated with FOLFIRI (fluorouracil, leucovorin, and irinotecan), XELOX (capecitabine and oxaliplatin), cetuximab and bevacizumab, and then received the next generation sequencing (NGS) and whose metastatic subcutaneous nodule was resected to generate patient-derived xenograft (PDX) models. Irinotecan 151-161 B-Raf proto-oncogene, serine/threonine kinase Homo sapiens 47-51 31045514-4 2019 OBJECTIVE: Investigation of the association between rs4759314 HOTAIR and rs3200401 MALAT1 polymorphisms and irinotecan-based chemotherapy in terms of drug efficacy and toxicity. Irinotecan 108-118 metastasis associated lung adenocarcinoma transcript 1 Homo sapiens 83-89 30396915-5 2019 Under these suppression conditions, cER values for irinotecan and topotecan (dual substrates of ABCB1 and ABCG2) were elevated by more than 4-fold and 2-fold, respectively, compared with cER values without the suppression. Irinotecan 51-61 ATP binding cassette subfamily B member 1 Canis lupus familiaris 96-101 29932028-4 2019 SN-38 is detoxified by the formation of SN-38 glucuronide, through UGT1A1. Irinotecan 0-5 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 67-73 29932028-5 2019 Genetic polymorphisms in the UGT1A1 gene are associated to higher exposures to SN-38 and severe toxicity. Irinotecan 79-84 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 29-35 30396915-5 2019 Under these suppression conditions, cER values for irinotecan and topotecan (dual substrates of ABCB1 and ABCG2) were elevated by more than 4-fold and 2-fold, respectively, compared with cER values without the suppression. Irinotecan 51-61 ATP binding cassette subfamily G member 2 Canis lupus familiaris 106-111 30595211-0 2019 Cytoprotective and genotoxic effects of vitamins K1 and B1 on irinotecan in vitro. Irinotecan 62-72 keratin 1 Homo sapiens 49-58 30372160-5 2019 The results showed that 5 mug/ml of OED combined with 40 muM of irinotecan possessed significant synergetic inhibition on SW-480 cells with a combination index (CI) of 0.56. Irinotecan 64-74 Oregon eye disease Homo sapiens 36-39 30372160-6 2019 Besides, the percentage of apoptotic cells was significantly increased from 69.57% (40 muM of irinotecan) or 72.7% (5 mug/ml of OED) to 95.6% after treatment of OED combined with irinotecan (OCI), suggesting OED and irinotecan possess the synergistic apoptotic effect (P < 0.01). Irinotecan 94-104 Oregon eye disease Homo sapiens 161-164 30372160-6 2019 Besides, the percentage of apoptotic cells was significantly increased from 69.57% (40 muM of irinotecan) or 72.7% (5 mug/ml of OED) to 95.6% after treatment of OED combined with irinotecan (OCI), suggesting OED and irinotecan possess the synergistic apoptotic effect (P < 0.01). Irinotecan 94-104 Oregon eye disease Homo sapiens 161-164 30372160-6 2019 Besides, the percentage of apoptotic cells was significantly increased from 69.57% (40 muM of irinotecan) or 72.7% (5 mug/ml of OED) to 95.6% after treatment of OED combined with irinotecan (OCI), suggesting OED and irinotecan possess the synergistic apoptotic effect (P < 0.01). Irinotecan 179-189 Oregon eye disease Homo sapiens 128-131 30372160-6 2019 Besides, the percentage of apoptotic cells was significantly increased from 69.57% (40 muM of irinotecan) or 72.7% (5 mug/ml of OED) to 95.6% after treatment of OED combined with irinotecan (OCI), suggesting OED and irinotecan possess the synergistic apoptotic effect (P < 0.01). Irinotecan 179-189 Oregon eye disease Homo sapiens 128-131 30372160-9 2019 These data indicated that OED could enhance antiproliferative effects of irinotecan on colorectal cancer cells, which was related with induction of apoptosis and regulations of activity of caspase-3. Irinotecan 73-83 Oregon eye disease Homo sapiens 26-29 28963609-6 2019 TIMP-1 overexpressing U87MG cells were significantly more resistant than low TIMP-1 expressing clones and parental cells when exposed to SN-38 (TOP1 inhibitor) or epirubicin (TOP2 inhibitor). Irinotecan 137-142 TIMP metallopeptidase inhibitor 1 Homo sapiens 0-6 30527657-1 2019 Differences in drug metabolism associated with UGT1A1 polymorphism could result in individualized local response to hepatic chemoembolization with irinotecan-eluting beads (DEBIRI) or predictable toxicities. Irinotecan 147-157 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 47-53 30009852-4 2018 Herein we evidenced, for the first time, that PTHrP induces resistance to CPT-11 in Caco-2 and HCT116 cells; although both cell lines responded to the drug through different molecular mechanisms, the chemoresistance by PTHrP in these models is mediated through ERK, which in turn is activated by PCK, Src and Akt. Irinotecan 74-80 parathyroid hormone like hormone Homo sapiens 46-51 28963609-11 2019 Whether TIMP-1 plays a role in irinotecan resistance and has a predictive potential in glioblastoma patients remains to be elucidated. Irinotecan 31-41 TIMP metallopeptidase inhibitor 1 Homo sapiens 8-14 30391354-0 2019 Pharmacological inhibition of bacterial beta-glucuronidase prevents irinotecan-induced diarrhea without impairing its antitumor efficacy in vivo. Irinotecan 68-78 glucuronidase, beta Mus musculus 40-58 30391354-2 2019 The underlying mechanism has been shown that the active metabolite of CPT-11, SN-38, is metabolized to the inactive metabolite SN-38 glucuronide (SN-38 G) during hepatic glucuronidation, and subsequently is exported into the intestine, where SN-38 G is hydrolyzed by bacterial beta-glucuronidase (betaG) to be SN-38, thus leading to intestinal toxicity. Irinotecan 70-76 glucuronidase, beta Mus musculus 277-295 30391354-2 2019 The underlying mechanism has been shown that the active metabolite of CPT-11, SN-38, is metabolized to the inactive metabolite SN-38 glucuronide (SN-38 G) during hepatic glucuronidation, and subsequently is exported into the intestine, where SN-38 G is hydrolyzed by bacterial beta-glucuronidase (betaG) to be SN-38, thus leading to intestinal toxicity. Irinotecan 78-83 glucuronidase, beta Mus musculus 277-295 30391354-2 2019 The underlying mechanism has been shown that the active metabolite of CPT-11, SN-38, is metabolized to the inactive metabolite SN-38 glucuronide (SN-38 G) during hepatic glucuronidation, and subsequently is exported into the intestine, where SN-38 G is hydrolyzed by bacterial beta-glucuronidase (betaG) to be SN-38, thus leading to intestinal toxicity. Irinotecan 127-132 glucuronidase, beta Mus musculus 277-295 30391354-2 2019 The underlying mechanism has been shown that the active metabolite of CPT-11, SN-38, is metabolized to the inactive metabolite SN-38 glucuronide (SN-38 G) during hepatic glucuronidation, and subsequently is exported into the intestine, where SN-38 G is hydrolyzed by bacterial beta-glucuronidase (betaG) to be SN-38, thus leading to intestinal toxicity. Irinotecan 127-132 glucuronidase, beta Mus musculus 277-295 30391354-2 2019 The underlying mechanism has been shown that the active metabolite of CPT-11, SN-38, is metabolized to the inactive metabolite SN-38 glucuronide (SN-38 G) during hepatic glucuronidation, and subsequently is exported into the intestine, where SN-38 G is hydrolyzed by bacterial beta-glucuronidase (betaG) to be SN-38, thus leading to intestinal toxicity. Irinotecan 127-132 glucuronidase, beta Mus musculus 277-295 30246377-0 2018 UGT1A1 polymorphisms in rectal cancer associated with the efficacy and toxicity of preoperative chemoradiotherapy using irinotecan. Irinotecan 120-130 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 29499902-9 2018 Furthermore, it was observed that those patients with wild-type in UGT family genes analysed have lower rates of toxicity associated with irinotecan treatment than those with certain mutated allele (P=.010). Irinotecan 138-148 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 67-70 29499902-10 2018 CONCLUSIONS: These results suggest that the presence of certain polymorphisms in the UGT1A family of genes is related to the development of toxicity during treatment with irinotecan. Irinotecan 171-181 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 85-90 30246377-11 2018 Our results indicate that UGT1A1 polymorphism is a predictive factor to determine the clinical efficacy of preoperative chemoradiotherapy and hematological toxicity induced by chemoradiotherapy using irinotecan in locally advanced rectal cancer patients. Irinotecan 200-210 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 26-32 29499902-0 2018 Effect of UGT, SLCO, ABCB and ABCC polymorphisms on irinotecan toxicity. Irinotecan 52-62 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 10-13 29499902-0 2018 Effect of UGT, SLCO, ABCB and ABCC polymorphisms on irinotecan toxicity. Irinotecan 52-62 ATP binding cassette subfamily C member 1 Homo sapiens 30-34 29499902-1 2018 BACKGROUND AND OBJECTIVES: Evaluate the relationship between the presence of polymorphisms in genes involved in the pharmacodynamics of irinotecan (UGT1A, SLCO1B1, ABCB1 and ABCC2) and the safety of irinotecan in the treatment of metastatic colorectal cancer (mCRC). Irinotecan 136-146 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 148-153 29499902-1 2018 BACKGROUND AND OBJECTIVES: Evaluate the relationship between the presence of polymorphisms in genes involved in the pharmacodynamics of irinotecan (UGT1A, SLCO1B1, ABCB1 and ABCC2) and the safety of irinotecan in the treatment of metastatic colorectal cancer (mCRC). Irinotecan 136-146 solute carrier organic anion transporter family member 1B1 Homo sapiens 155-162 29499902-1 2018 BACKGROUND AND OBJECTIVES: Evaluate the relationship between the presence of polymorphisms in genes involved in the pharmacodynamics of irinotecan (UGT1A, SLCO1B1, ABCB1 and ABCC2) and the safety of irinotecan in the treatment of metastatic colorectal cancer (mCRC). Irinotecan 136-146 ATP binding cassette subfamily B member 1 Homo sapiens 164-169 29499902-1 2018 BACKGROUND AND OBJECTIVES: Evaluate the relationship between the presence of polymorphisms in genes involved in the pharmacodynamics of irinotecan (UGT1A, SLCO1B1, ABCB1 and ABCC2) and the safety of irinotecan in the treatment of metastatic colorectal cancer (mCRC). Irinotecan 136-146 ATP binding cassette subfamily C member 2 Homo sapiens 174-179 30246377-1 2018 The purpose of the present study was to assess the efficacy and toxicity of preoperative chemoradiotherapy using irinotecan against locally advanced lower rectal cancer according to UDP-glucuronosyltransferase 1A1 (UGT1A1) polymorphisms. Irinotecan 113-123 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 182-213 30246377-1 2018 The purpose of the present study was to assess the efficacy and toxicity of preoperative chemoradiotherapy using irinotecan against locally advanced lower rectal cancer according to UDP-glucuronosyltransferase 1A1 (UGT1A1) polymorphisms. Irinotecan 113-123 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 215-221 30527181-2 2018 Uridine diphosphate (UDP)-glucuronosyltransferase 1A1 (UGT1A1) is thought to be largely responsible for the glucuronidation of etoposide as well as that of irinotecan, suggesting that polymorphisms of UGT1A1 might be predictive of etoposide toxicity. Irinotecan 156-166 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-53 30367935-0 2018 beta-Casein micelles for oral delivery of SN-38 and elacridar to overcome BCRP-mediated multidrug resistance in gastric cancer. Irinotecan 42-47 casein beta Homo sapiens 0-11 30367935-5 2018 The chemotherapeutic drug SN-38, a BCRP transport substrate, and the BCRP efflux transport inhibitor, elacridar, exhibited high binding affinity to beta-CN, as demonstrated by spectrophotometry and spectrofluorometry. Irinotecan 26-31 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 35-39 30367935-5 2018 The chemotherapeutic drug SN-38, a BCRP transport substrate, and the BCRP efflux transport inhibitor, elacridar, exhibited high binding affinity to beta-CN, as demonstrated by spectrophotometry and spectrofluorometry. Irinotecan 26-31 casein beta Homo sapiens 148-155 30191738-2 2018 TDP1 plays an essential role in the resistance of cancer cells to currently used antitumour drugs based on Top1 inhibitors such as topotecan and irinotecan. Irinotecan 145-155 tyrosyl-DNA phosphodiesterase 1 Homo sapiens 0-4 30692298-10 2018 Chemotherapy containing cisplatin(CDDP)and irinotecan(CPT-11)was initiated; although the blood hCG level was temporally lowered, the patient died of liver failure 8 months after the surgery. Irinotecan 43-53 hypertrichosis 2 (generalised, congenital) Homo sapiens 95-98 30692298-10 2018 Chemotherapy containing cisplatin(CDDP)and irinotecan(CPT-11)was initiated; although the blood hCG level was temporally lowered, the patient died of liver failure 8 months after the surgery. Irinotecan 54-60 hypertrichosis 2 (generalised, congenital) Homo sapiens 95-98 30527181-2 2018 Uridine diphosphate (UDP)-glucuronosyltransferase 1A1 (UGT1A1) is thought to be largely responsible for the glucuronidation of etoposide as well as that of irinotecan, suggesting that polymorphisms of UGT1A1 might be predictive of etoposide toxicity. Irinotecan 156-166 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 55-61 30527181-2 2018 Uridine diphosphate (UDP)-glucuronosyltransferase 1A1 (UGT1A1) is thought to be largely responsible for the glucuronidation of etoposide as well as that of irinotecan, suggesting that polymorphisms of UGT1A1 might be predictive of etoposide toxicity. Irinotecan 156-166 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 201-207 30139840-1 2018 LESSONS LEARNED: The maximum tolerated dose of the combination of linsitinib and irinotecan is linsitinib 450 mg daily on days 1-3 every 7 days and irinotecan 125 mg/m2 days 1 and 8 of a 21-day cycle.The adverse effects associated with the combination are not significantly increased beyond what is expected of each drug as a single agent.Multiple negative trials of insulin-like growth factor-1 receptor inhibitors performed in unselected patient populations led to the early discontinuation of linistinib development and this trial.Earlier integration of assessment of potential predictive biomarkers into clinical trials, as was planned in this study, is vital to the development of targeted therapies in oncology. Irinotecan 81-91 insulin like growth factor 1 receptor Homo sapiens 367-404 30538568-0 2018 Value of plasma SN-38 levels and DPD activity in irinotecan-based individualized chemotherapy for advanced colorectal cancer with heterozygous type UGT1A1*6 or UGT1A1*28. Irinotecan 49-59 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 148-154 30538568-0 2018 Value of plasma SN-38 levels and DPD activity in irinotecan-based individualized chemotherapy for advanced colorectal cancer with heterozygous type UGT1A1*6 or UGT1A1*28. Irinotecan 49-59 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 160-166 30498448-4 2018 Genetic variants resulting in decreased enzymatic activity of uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) and dihydropyrimidine dehydrogenase (DPD) are known to increase irinotecan and 5-fluorouracil-related toxicity, respectively. Irinotecan 183-193 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 62-109 30498448-4 2018 Genetic variants resulting in decreased enzymatic activity of uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) and dihydropyrimidine dehydrogenase (DPD) are known to increase irinotecan and 5-fluorouracil-related toxicity, respectively. Irinotecan 183-193 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 111-117 30498448-4 2018 Genetic variants resulting in decreased enzymatic activity of uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) and dihydropyrimidine dehydrogenase (DPD) are known to increase irinotecan and 5-fluorouracil-related toxicity, respectively. Irinotecan 183-193 dihydropyrimidine dehydrogenase Homo sapiens 123-154 30498448-4 2018 Genetic variants resulting in decreased enzymatic activity of uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) and dihydropyrimidine dehydrogenase (DPD) are known to increase irinotecan and 5-fluorouracil-related toxicity, respectively. Irinotecan 183-193 dihydropyrimidine dehydrogenase Homo sapiens 156-159 30385821-6 2018 Similar inhibition of pRAD50 (68%) was observed following ATM inhibitor treatment post irinotecan in a colorectal cancer xenograft model. Irinotecan 87-97 ATM serine/threonine kinase Homo sapiens 58-61 30396964-0 2018 Patients with hMLH1 or/and hMSH2-deficient Metastatic Colorectal Cancer Are Associated with Reduced Levels of Vascular Endothelial Growth Factor-1 Expression and Higher Response Rate to Irinotecan-based Regimen. Irinotecan 186-196 mutL homolog 1 Homo sapiens 14-19 30519050-0 2018 Association of MLH1 single nucleotide polymorphisms with clinical outcomes of first-line irinotecan-based chemotherapy in colorectal cancer. Irinotecan 89-99 mutL homolog 1 Homo sapiens 15-19 30519050-2 2018 The aim of this study explored the influence of MLH1 SNPs on clinical outcomes of first-line irinotecan-based chemotherapy in CRC. Irinotecan 93-103 mutL homolog 1 Homo sapiens 48-52 30519050-11 2018 Conclusion: The AA variant of MLH1 rs1800734 SNPs has a longer PFS in first-line irinotecan-based chemotherapy for mCRC patients, and the result needs to be further confirmed by prospective studies in the future. Irinotecan 81-91 mutL homolog 1 Homo sapiens 30-34 30257334-3 2018 The MTT assay was used to detect growth inhibition of differentially-expressed KDM5c colon cancer cells, for which L-OHP or CPT-11 were added. Irinotecan 124-130 lysine demethylase 5C Homo sapiens 79-84 30135242-3 2018 The hiPSC-IECs showed the transport activities of P-glycoprotein (P-gp), breast cancer resistance protein (BCRP), and peptide transporter 1 (PEPT1), revealed by using their probe substrates ([3H]digoxin, sulfasalazine, and [14C]glycylsarcosine), and the metabolic activities of CYP3A4, CES2, and CES1, which were clarified using their probe substrates (midazolam, irinotecan, and temocapril). Irinotecan 364-374 ATP binding cassette subfamily B member 1 Homo sapiens 50-64 30135242-3 2018 The hiPSC-IECs showed the transport activities of P-glycoprotein (P-gp), breast cancer resistance protein (BCRP), and peptide transporter 1 (PEPT1), revealed by using their probe substrates ([3H]digoxin, sulfasalazine, and [14C]glycylsarcosine), and the metabolic activities of CYP3A4, CES2, and CES1, which were clarified using their probe substrates (midazolam, irinotecan, and temocapril). Irinotecan 364-374 ATP binding cassette subfamily B member 1 Homo sapiens 66-70 30166404-0 2018 Gene-by-Environment Interaction of Bcrp-/- and Methionine- and Choline-Deficient Diet-Induced Nonalcoholic Steatohepatitis Alters SN-38 Disposition. Irinotecan 130-135 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 35-39 30166404-4 2018 SN-38, the active irinotecan metabolite, is reported to be a substrate for Bcrp, whereas SN-38 glucuronide (SN-38G) is a Mrp2 substrate. Irinotecan 0-5 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 75-79 30135242-3 2018 The hiPSC-IECs showed the transport activities of P-glycoprotein (P-gp), breast cancer resistance protein (BCRP), and peptide transporter 1 (PEPT1), revealed by using their probe substrates ([3H]digoxin, sulfasalazine, and [14C]glycylsarcosine), and the metabolic activities of CYP3A4, CES2, and CES1, which were clarified using their probe substrates (midazolam, irinotecan, and temocapril). Irinotecan 364-374 solute carrier family 15 member 1 Homo sapiens 118-139 30166404-4 2018 SN-38, the active irinotecan metabolite, is reported to be a substrate for Bcrp, whereas SN-38 glucuronide (SN-38G) is a Mrp2 substrate. Irinotecan 18-28 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 75-79 30166404-4 2018 SN-38, the active irinotecan metabolite, is reported to be a substrate for Bcrp, whereas SN-38 glucuronide (SN-38G) is a Mrp2 substrate. Irinotecan 89-94 ATP binding cassette subfamily C member 2 Rattus norvegicus 121-125 30166404-5 2018 The purpose of this study was to determine the function of Bcrp in NASH through alterations in the disposition of SN-38 and SN-38G in a Bcrp knockout (Bcrp-/- KO) and methionine- and choline-deficient (MCD) model of NASH. Irinotecan 114-119 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 59-63 30135242-3 2018 The hiPSC-IECs showed the transport activities of P-glycoprotein (P-gp), breast cancer resistance protein (BCRP), and peptide transporter 1 (PEPT1), revealed by using their probe substrates ([3H]digoxin, sulfasalazine, and [14C]glycylsarcosine), and the metabolic activities of CYP3A4, CES2, and CES1, which were clarified using their probe substrates (midazolam, irinotecan, and temocapril). Irinotecan 364-374 solute carrier family 15 member 1 Homo sapiens 141-146 30166404-9 2018 These data indicate that Bcrp is not solely responsible for SN-38 biliary efflux, but rather implicate a combined role for BCRP and MRP2. Irinotecan 60-65 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 25-29 30166404-10 2018 Furthermore, the disposition of SN-38 and SN-38G is altered by Bcrp-/- and NASH in a gene-by-environment interaction and may result in variable drug response to irinotecan therapy in polymorphic patients. Irinotecan 32-37 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 63-67 30361780-0 2018 Irinotecan Alters the Disposition of Morphine Via Inhibition of Organic Cation Transporter 1 (OCT1) and 2 (OCT2). Irinotecan 0-10 solute carrier family 22 member 1 Homo sapiens 64-92 30166404-10 2018 Furthermore, the disposition of SN-38 and SN-38G is altered by Bcrp-/- and NASH in a gene-by-environment interaction and may result in variable drug response to irinotecan therapy in polymorphic patients. Irinotecan 42-47 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 63-67 30166404-10 2018 Furthermore, the disposition of SN-38 and SN-38G is altered by Bcrp-/- and NASH in a gene-by-environment interaction and may result in variable drug response to irinotecan therapy in polymorphic patients. Irinotecan 161-171 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 63-67 30360603-0 2018 Effects of ABCC2 and SLCO1B1 Polymorphisms on Treatment Responses in Thai Metastatic Colorectal Cancer Patients Treated with Irinotecan-Based Chemotherapy Purpose: Irinotecan is an anticancer medicine which is used mostly in metastatic colorectal cancer (mCRC) treatmentas second or third line chemotherapy. Irinotecan 164-174 ATP binding cassette subfamily C member 2 Homo sapiens 11-16 30360603-0 2018 Effects of ABCC2 and SLCO1B1 Polymorphisms on Treatment Responses in Thai Metastatic Colorectal Cancer Patients Treated with Irinotecan-Based Chemotherapy Purpose: Irinotecan is an anticancer medicine which is used mostly in metastatic colorectal cancer (mCRC) treatmentas second or third line chemotherapy. Irinotecan 164-174 solute carrier organic anion transporter family member 1B1 Homo sapiens 21-28 30361780-0 2018 Irinotecan Alters the Disposition of Morphine Via Inhibition of Organic Cation Transporter 1 (OCT1) and 2 (OCT2). Irinotecan 0-10 solute carrier family 22 member 1 Homo sapiens 94-98 30361780-0 2018 Irinotecan Alters the Disposition of Morphine Via Inhibition of Organic Cation Transporter 1 (OCT1) and 2 (OCT2). Irinotecan 0-10 POU class 2 homeobox 2 Homo sapiens 107-111 30120489-0 2018 Replica to the Opinion Letter regarding the article "Sensitization of colorectal cancer cells to irinotecan by the Survivin inhibitor LLP3 depends on XAF1 proficiency in the context of mutated p53" (Arch Toxicol https://doi.org/10.1007/s00204-018-240-x). Irinotecan 97-107 XIAP associated factor 1 Homo sapiens 150-154 30361780-7 2018 All of the six drugs examined, including amitriptyline, fluoxetine, imipramine, irinotecan, ondansetron, and verapamil, were inhibitors of OCT1/2-mediated morphine uptake. Irinotecan 80-90 solute carrier family 22 member 1 Homo sapiens 139-143 30361780-10 2018 Clinician should be aware that the disposition of and thus the response to morphine may be altered by co-administration of an OCT1/2 inhibitor, such as irinotecan. Irinotecan 152-162 solute carrier family 22 member 1 Homo sapiens 126-132 32913994-0 2018 Temporal Dynamics of Genomic Alterations in a BRCA1 Germline-Mutated Pancreatic Cancer With Low Genomic Instability Burden but Exceptional Response to Fluorouracil, Oxaliplatin, Leucovorin, and Irinotecan. Irinotecan 194-204 BRCA1 DNA repair associated Homo sapiens 46-51 29753314-5 2018 In contrast, both types of mGluR8 activators significantly enhanced toxic effects of doxorubicin and irinotecan in RA-SH-SY5Y cells. Irinotecan 101-111 glutamate receptor, metabotropic 8 Mus musculus 27-33 30120489-0 2018 Replica to the Opinion Letter regarding the article "Sensitization of colorectal cancer cells to irinotecan by the Survivin inhibitor LLP3 depends on XAF1 proficiency in the context of mutated p53" (Arch Toxicol https://doi.org/10.1007/s00204-018-240-x). Irinotecan 97-107 tumor protein p53 Homo sapiens 193-196 30213564-0 2018 Clinical utility of ABCB1 genotyping for preventing toxicity in treatment with irinotecan. Irinotecan 79-89 ATP binding cassette subfamily B member 1 Homo sapiens 20-25 29932297-0 2018 UGT1A1*6 and UGT1A1*28 polymorphisms are correlated with irinotecan-induced toxicity: A meta-analysis. Irinotecan 57-67 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 29932297-0 2018 UGT1A1*6 and UGT1A1*28 polymorphisms are correlated with irinotecan-induced toxicity: A meta-analysis. Irinotecan 57-67 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 13-19 29932297-1 2018 BACKGROUND: Previous articles explored the role of UGT1A1 polymorphism on predicting irinotecan-induced toxicity, but the conclusions were still inconsistent and not comprehensive. Irinotecan 85-95 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 51-57 29932297-2 2018 We performed this meta-analysis to investigate the association between UGT1A1 polymorphism and irinotecan-induced toxicity. Irinotecan 95-105 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 71-77 29932297-8 2018 Both UGT1A1*6 and UGT1A1*28 polymorphism are significantly associated with severe irinotecan-induced toxicity. Irinotecan 82-92 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 5-11 29932297-8 2018 Both UGT1A1*6 and UGT1A1*28 polymorphism are significantly associated with severe irinotecan-induced toxicity. Irinotecan 82-92 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 18-24 29932297-12 2018 Subgroup analysis exhibited that for UGT1A1*6 polymorphism, patients treated with low-dose irinotecan were at a notable risk of toxicity. Irinotecan 91-101 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 37-43 29932297-13 2018 Moreover, the association between UGT1A1*6 polymorphism and irinotecan-induced toxicity was found in patients suffering from respiratory system cancers. Irinotecan 60-70 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 34-40 29932297-14 2018 CONCLUSIONS: Both UGT1A1*6 and UGT1A1*28 polymorphisms can be considered as predictors of irinotecan-induced toxicity, with effect varying by race, cancer type and irinotecan dose. Irinotecan 90-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 18-24 29932297-14 2018 CONCLUSIONS: Both UGT1A1*6 and UGT1A1*28 polymorphisms can be considered as predictors of irinotecan-induced toxicity, with effect varying by race, cancer type and irinotecan dose. Irinotecan 90-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 31-37 30069943-13 2018 CONCLUSIONS: A novel ADAM17 inhibitor ZLDI-8 may be a potential chemosensitizer which sensitized CRC cells to 5-fluorouracil or irinotecan by reversing Notch and EMT pathways. Irinotecan 128-138 ADAM metallopeptidase domain 17 Homo sapiens 21-27 30130528-7 2018 Irinotecan treatment induced elevation of aspartate aminotransferase (AST) in rats without diarrheal symptoms and without an increase in circulating pro-inflammatory mediators. Irinotecan 0-10 glutamic-oxaloacetic transaminase 2 Rattus norvegicus 42-68 30130528-7 2018 Irinotecan treatment induced elevation of aspartate aminotransferase (AST) in rats without diarrheal symptoms and without an increase in circulating pro-inflammatory mediators. Irinotecan 0-10 glutamic-oxaloacetic transaminase 2 Rattus norvegicus 70-73 30085332-0 2018 Radiosensitization by irinotecan is attributed to G2/M phase arrest, followed by enhanced apoptosis, probably through the ATM/Chk/Cdc25C/Cdc2 pathway in p53-mutant colorectal cancer cells. Irinotecan 22-32 ATM serine/threonine kinase Homo sapiens 122-125 30085332-0 2018 Radiosensitization by irinotecan is attributed to G2/M phase arrest, followed by enhanced apoptosis, probably through the ATM/Chk/Cdc25C/Cdc2 pathway in p53-mutant colorectal cancer cells. Irinotecan 22-32 choline kinase alpha Homo sapiens 126-129 30085332-0 2018 Radiosensitization by irinotecan is attributed to G2/M phase arrest, followed by enhanced apoptosis, probably through the ATM/Chk/Cdc25C/Cdc2 pathway in p53-mutant colorectal cancer cells. Irinotecan 22-32 cell division cycle 25C Homo sapiens 130-136 30085332-0 2018 Radiosensitization by irinotecan is attributed to G2/M phase arrest, followed by enhanced apoptosis, probably through the ATM/Chk/Cdc25C/Cdc2 pathway in p53-mutant colorectal cancer cells. Irinotecan 22-32 cyclin dependent kinase 1 Homo sapiens 130-134 30085332-0 2018 Radiosensitization by irinotecan is attributed to G2/M phase arrest, followed by enhanced apoptosis, probably through the ATM/Chk/Cdc25C/Cdc2 pathway in p53-mutant colorectal cancer cells. Irinotecan 22-32 tumor protein p53 Homo sapiens 153-156 30085332-4 2018 In the present study, we examined the radiosensitizing effects of irinotecan on the p53-mutant colorectal cancer cell lines, HT29 and SW620, and explored the potential underlying mechanisms. Irinotecan 66-76 tumor protein p53 Homo sapiens 84-87 30085332-11 2018 In addition, the expression of Ser216p-Cdc25C was also increased in the combined group, indicating that irinotecan likely radiosensitized the p53-mutant HT29 and SW620 cells through the ATM/Chk/Cdc25C/Cdc2 pathway. Irinotecan 104-114 cell division cycle 25C Homo sapiens 39-45 30085332-11 2018 In addition, the expression of Ser216p-Cdc25C was also increased in the combined group, indicating that irinotecan likely radiosensitized the p53-mutant HT29 and SW620 cells through the ATM/Chk/Cdc25C/Cdc2 pathway. Irinotecan 104-114 tumor protein p53 Homo sapiens 142-145 30085332-11 2018 In addition, the expression of Ser216p-Cdc25C was also increased in the combined group, indicating that irinotecan likely radiosensitized the p53-mutant HT29 and SW620 cells through the ATM/Chk/Cdc25C/Cdc2 pathway. Irinotecan 104-114 ATM serine/threonine kinase Homo sapiens 186-189 30085332-11 2018 In addition, the expression of Ser216p-Cdc25C was also increased in the combined group, indicating that irinotecan likely radiosensitized the p53-mutant HT29 and SW620 cells through the ATM/Chk/Cdc25C/Cdc2 pathway. Irinotecan 104-114 choline kinase alpha Homo sapiens 190-193 30085332-11 2018 In addition, the expression of Ser216p-Cdc25C was also increased in the combined group, indicating that irinotecan likely radiosensitized the p53-mutant HT29 and SW620 cells through the ATM/Chk/Cdc25C/Cdc2 pathway. Irinotecan 104-114 cell division cycle 25C Homo sapiens 194-200 30085332-11 2018 In addition, the expression of Ser216p-Cdc25C was also increased in the combined group, indicating that irinotecan likely radiosensitized the p53-mutant HT29 and SW620 cells through the ATM/Chk/Cdc25C/Cdc2 pathway. Irinotecan 104-114 cyclin dependent kinase 1 Homo sapiens 39-43 28747102-1 2018 We report the case of a 50-year-old human immunodeficiency virus-positive patient with stage IV KRAS-mutated colorectal cancer who experienced visible muscle twitching in the right lateral triceps brachii from irinotecan administration for which typical supportive care measures were unsuccessful, including the administration of atropine and slowing down the infusion rate. Irinotecan 210-220 KRAS proto-oncogene, GTPase Homo sapiens 96-100 29273261-1 2018 PURPOSE: We aimed to identify the maximum tolerated dose (MTD) of weekly irinotecan in combination with capecitabine-based neoadjuvant chemoradiation according to the UGT1A1*28 genotype in patients with locally advanced rectal cancer. Irinotecan 73-83 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 167-173 29273261-11 2018 CONCLUSION: A higher dose of weekly irinotecan in combination with capecitabine-based CRT is feasible under the guidance of the UGT1A1*28 genotype. Irinotecan 36-46 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 128-134 30213564-9 2018 Genotyping of ABCB1 variants can help to prevent severe adverse reactions to irinotecan-based treatments in colorectal cancer. Irinotecan 77-87 ATP binding cassette subfamily B member 1 Homo sapiens 14-19 29034773-10 2018 In contrast, afatinib stimulated OATP1B1-mediated uptake of FL and SN-38, ceritinib stimulated that of valsartan, and nintedanib stimulated that of FL and valsartan. Irinotecan 67-72 solute carrier organic anion transporter family member 1B1 Homo sapiens 33-40 29034773-6 2018 We used OATP1B1-transfected cells to compare the effects of molecular-targeted agents on OATP1B1-mediated uptake of fluorescein (FL), 2",7"-dichlorofluorescein (DCF), atorvastatin, SN-38 and valsartan. Irinotecan 181-186 solute carrier organic anion transporter family member 1B1 Homo sapiens 89-96 28229371-5 2018 RESULTS: In patients whose tumors expressed the KRAS wild type, the longest median OS was observed with irinotecan-based chemotherapy combined with bevacizumab (38 months), or with cetuximab (41 months). Irinotecan 104-114 KRAS proto-oncogene, GTPase Homo sapiens 48-52 29980022-3 2018 The intrinsic fluorescence of SN-38 allowed its quantification in the range 10-500 ng mL-1 with a LOQ of 5.0 ng mL-1 and a LOD of 1.5 ng mL-1. Irinotecan 30-35 L1 cell adhesion molecule Mus musculus 86-90 29980022-3 2018 The intrinsic fluorescence of SN-38 allowed its quantification in the range 10-500 ng mL-1 with a LOQ of 5.0 ng mL-1 and a LOD of 1.5 ng mL-1. Irinotecan 30-35 L1 cell adhesion molecule Mus musculus 112-116 29980022-3 2018 The intrinsic fluorescence of SN-38 allowed its quantification in the range 10-500 ng mL-1 with a LOQ of 5.0 ng mL-1 and a LOD of 1.5 ng mL-1. Irinotecan 30-35 L1 cell adhesion molecule Mus musculus 112-116 29885518-4 2018 The mGluR8-downregulated SH-SY5Y clones proliferated faster and were more resistant to cytotoxic action of staurosporine, doxorubicin, irinotecan and cisplatin when compared to control cells. Irinotecan 135-145 glutamate receptor, metabotropic 8 Mus musculus 4-10 29885518-6 2018 The mGluR8-downregulated LN229 clones migrated faster and were less prone to cell-damaging effect of staurosporine and irinotecan when compared with relevant control cells. Irinotecan 119-129 glutamate receptor, metabotropic 8 Mus musculus 4-10 30075623-5 2018 Introduction of a 6-nitro function led to extraordinarily potent compounds that were highly selective for ABCG2 and also able to reverse the MDR toward the chemotherapeutic drugs SN-38 and mitoxantrone. Irinotecan 179-184 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 106-111 29860358-0 2018 Temozolomide and irinotecan (TEMIRI regimen) as salvage treatment of irinotecan-sensitive advanced colorectal cancer patients bearing MGMT methylation. Irinotecan 17-27 O-6-methylguanine-DNA methyltransferase Homo sapiens 134-138 30103709-6 2018 RESULTS: Irinotecan induced in vivo the activation of AKT and MEK1 phosphorylation. Irinotecan 9-19 thymoma viral proto-oncogene 1 Mus musculus 54-57 30103709-6 2018 RESULTS: Irinotecan induced in vivo the activation of AKT and MEK1 phosphorylation. Irinotecan 9-19 mitogen-activated protein kinase kinase 1 Mus musculus 62-66 30103709-7 2018 The dose matrix approach showed that BKM120 (PI3K inhibitor) and MEK162 (MEK inhibitor) are synergistic in vitro and in vivo with a cytostatic and cytotoxic effect, while combination of BKM120 and irinotecan or MEK162 and irinotecan are only additive or even antagonistic. Irinotecan 197-207 midkine Mus musculus 65-68 30103709-7 2018 The dose matrix approach showed that BKM120 (PI3K inhibitor) and MEK162 (MEK inhibitor) are synergistic in vitro and in vivo with a cytostatic and cytotoxic effect, while combination of BKM120 and irinotecan or MEK162 and irinotecan are only additive or even antagonistic. Irinotecan 222-232 midkine Mus musculus 65-68 29925895-0 2018 Potential role of PIN1 genotypes in predicting benefit from oxaliplatin-based and irinotecan-based treatment in patients with metastatic colorectal cancer. Irinotecan 82-92 peptidylprolyl cis/trans isomerase, NIMA-interacting 1 Homo sapiens 18-22 29925895-2 2018 We hypothesized that genetic polymorphisms in PIN1-related pathways may affect tumor sensitivity to oxaliplatin or irinotecan in metastatic colorectal cancer (mCRC) patients. Irinotecan 115-125 peptidylprolyl cis/trans isomerase, NIMA-interacting 1 Homo sapiens 46-50 29925895-9 2018 Germline PIN1 polymorphisms may predict clinical outcomes in mCRC patients receiving oxaliplatin-based or irinotecan-based therapy, and identify specific populations favorable to oxaliplatin plus irinotecan combination therapy. Irinotecan 106-116 peptidylprolyl cis/trans isomerase, NIMA-interacting 1 Homo sapiens 9-13 29925895-9 2018 Germline PIN1 polymorphisms may predict clinical outcomes in mCRC patients receiving oxaliplatin-based or irinotecan-based therapy, and identify specific populations favorable to oxaliplatin plus irinotecan combination therapy. Irinotecan 196-206 peptidylprolyl cis/trans isomerase, NIMA-interacting 1 Homo sapiens 9-13 30139029-0 2018 [Relationship between UGT1A1 gene polymorphisms and irinotecan-induced severe adverse events]. Irinotecan 52-62 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 22-28 30139029-1 2018 Objective: To investigate the relationship between UGT1A1*6, UGT1A1*28, UGT1A1*60 and UGT1A1*93 polymorphisms and irinotecan-induced severe adverse reactions(grade 3-4 delayed diarrhea and neutropenia) in Chinese cancer patients. Irinotecan 114-124 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 51-57 30139029-16 2018 Conclusion: The cancer patients who carried UGT1A1*6, UGT1A1*28 and UGT1A1*60 gene polymorphisms have high risk of severe adverse events caused by irinotecan-based chemotherapy. Irinotecan 147-157 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 44-50 30139029-16 2018 Conclusion: The cancer patients who carried UGT1A1*6, UGT1A1*28 and UGT1A1*60 gene polymorphisms have high risk of severe adverse events caused by irinotecan-based chemotherapy. Irinotecan 147-157 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 54-60 30139029-16 2018 Conclusion: The cancer patients who carried UGT1A1*6, UGT1A1*28 and UGT1A1*60 gene polymorphisms have high risk of severe adverse events caused by irinotecan-based chemotherapy. Irinotecan 147-157 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 54-60 30117422-1 2018 The adverse reaction to irinotecan is related to the single nucleotide polymorphism (SNP) of UGT1A1*6 genotype. Irinotecan 24-34 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-99 30111719-1 2018 As part of our initial efforts into developing a tumor-targeting therapy, C-10 substituted derivatives of a camptothecin analog (SN-38) have been synthesized (2-, 3- and 4-nitrobenzyl) for use as potential hypoxia-activated prodrugs and evaluated for their cytotoxicity, topoisomerase I inhibition and electrochemical (reductive) properties. Irinotecan 129-134 homeobox C10 Homo sapiens 74-78 30111719-7 2018 Chemical reduction of the 4-nitrobenzyl analog led to the formation/release of SN-38 and validated the prodrug ability of the C-10 substituted derivative. Irinotecan 79-84 homeobox C10 Homo sapiens 126-130 30016861-2 2018 Abnormal CE2 levels are associated with various cancers, and CE2 is a key mediator of anticancer prodrugs, including irinotecan. Irinotecan 117-127 carboxylesterase 2H Mus musculus 61-64 30016861-6 2018 In CE2 overexpression studies, cancer cells had a markedly enhanced sensitivity to the cytotoxic effect of irinotecan, corresponding well with the TPM ratio of the probe. Irinotecan 107-117 carboxylesterase 2H Mus musculus 3-6 29980405-5 2018 Upon introduction of the WT-TP53 gene into the MIA-PaCa-2 pancreatic cancer cell line, the sensitivity to drugs used to treat pancreatic cancer cells such as: gemcitabine, fluorouracil (5FU), cisplatin, irinotecan, oxaliplatin, and paclitaxel increased significantly. Irinotecan 203-213 tumor protein p53 Homo sapiens 28-32 29947891-0 2018 Sensitization of colorectal cancer cells to irinotecan by the Survivin inhibitor LLP3 depends on XAF1 proficiency in the context of mutated p53. Irinotecan 44-54 XIAP associated factor 1 Homo sapiens 97-101 29947891-0 2018 Sensitization of colorectal cancer cells to irinotecan by the Survivin inhibitor LLP3 depends on XAF1 proficiency in the context of mutated p53. Irinotecan 44-54 tumor protein p53 Homo sapiens 140-143 29947891-7 2018 When combined with irinotecan, expression of the tumor suppressor X-linked inhibitor of apoptosis factor 1 (XAF1) plays a decisive role for sensitization of CRC cells to this first-line drug, however, only in the p53-mutated background. Irinotecan 19-29 XIAP associated factor 1 Homo sapiens 108-112 29860358-0 2018 Temozolomide and irinotecan (TEMIRI regimen) as salvage treatment of irinotecan-sensitive advanced colorectal cancer patients bearing MGMT methylation. Irinotecan 69-79 O-6-methylguanine-DNA methyltransferase Homo sapiens 134-138 29860358-2 2018 In this phase II trial, irinotecan and temozolomide (TEMIRI) combination regimen was assessed in irinotecan-sensitive, MGMT methylated/microsatellite stable (MSS) pretreated mCRC patients. Irinotecan 24-34 O-6-methylguanine-DNA methyltransferase Homo sapiens 119-123 29860358-14 2018 Conclusions: TEMIRI regimen is a safe and active option in pre-treated, irinotecan-sensitive mCRC patients with MGMT methylation. Irinotecan 72-82 O-6-methylguanine-DNA methyltransferase Homo sapiens 112-116 29721577-6 2018 Results showed that CPT-11 caused dramatic changes in heart tissue for oxidative stress parameters (TBARS, SOD, CAT, GSH, and GPx levels), histological tissue damage, and cytokine levels (TNF and IL-4). Irinotecan 20-26 tumor necrosis factor Rattus norvegicus 188-191 28679068-0 2018 Berberine hydrochloride counteracts enhanced IL-8 expression induced by SN 38 in AGS cells. Irinotecan 72-77 C-X-C motif chemokine ligand 8 Homo sapiens 45-49 28679068-3 2018 BER could down-regulate the enhanced IL-8 expression through down-regulating ERK1/2 and p38 MAPK over-activation induced by SN 38. Irinotecan 124-129 C-X-C motif chemokine ligand 8 Homo sapiens 37-41 28679068-3 2018 BER could down-regulate the enhanced IL-8 expression through down-regulating ERK1/2 and p38 MAPK over-activation induced by SN 38. Irinotecan 124-129 mitogen-activated protein kinase 3 Homo sapiens 77-83 28679068-4 2018 The increased IL-8 mediated adhesive ability of AGS cells to HUVECs induced by SN 38, could be reduced by BER. Irinotecan 79-84 C-X-C motif chemokine ligand 8 Homo sapiens 14-18 29908105-0 2018 Phase II trial of biweekly cetuximab and irinotecan as third-line therapy for pretreated KRAS exon 2 wild-type colorectal cancer. Irinotecan 41-51 KRAS proto-oncogene, GTPase Homo sapiens 89-93 29909091-2 2018 We assessed potential clinical variables that may predict toxicity and more specifically the role of UGT1A1 polymorphisms associated with irinotecan toxicity. Irinotecan 138-148 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 101-107 29721577-6 2018 Results showed that CPT-11 caused dramatic changes in heart tissue for oxidative stress parameters (TBARS, SOD, CAT, GSH, and GPx levels), histological tissue damage, and cytokine levels (TNF and IL-4). Irinotecan 20-26 interleukin 4 Rattus norvegicus 196-200 30013661-1 2018 Transarterial chemoembolization with irinotecan loaded beads (DEBIRI-TACE) represents an investigative treatment option for patients with metastatic colorectal cancer (mCRC). Irinotecan 37-47 ADAM metallopeptidase domain 17 Homo sapiens 69-73 29533113-1 2018 BACKGROUND: Data on temozolomide (TEM) and irinotecan (IRI) activity in recurrent Ewing sarcoma (EWS), especially in adult patients, are limited. Irinotecan 43-53 EWS RNA binding protein 1 Homo sapiens 97-100 30140379-0 2018 Topoisomerase I inhibitor, irinotecan, depletes regulatory T cells and up-regulates MHC class I and PD-L1 expression, resulting in a supra-additive antitumor effect when combined with anti-PD-L1 antibodies. Irinotecan 27-37 CD274 antigen Mus musculus 100-105 30140379-0 2018 Topoisomerase I inhibitor, irinotecan, depletes regulatory T cells and up-regulates MHC class I and PD-L1 expression, resulting in a supra-additive antitumor effect when combined with anti-PD-L1 antibodies. Irinotecan 27-37 CD274 antigen Mus musculus 189-194 30140379-4 2018 Despite a transient decrease in lymphocytes in the peripheral blood after irinotecan treatment, the antitumor activity of anti-PD-L1 antibody plus irinotecan was significantly greater than each agent alone. Irinotecan 74-84 CD274 antigen Mus musculus 127-132 30140379-7 2018 In addition, irinotecan augmented MHC class I expression on tumor cells and concurrently increased PD-L1 expression on tumor cells and tumor-infiltrating immune cells. Irinotecan 13-23 CD274 antigen Mus musculus 99-104 30140379-8 2018 These results indicate that irinotecan may enhance the effect of T cell activation caused by anti-PD-L1 treatment by reducing Tregs and augmenting MHC class I-mediated tumor antigen presentation, and concurrent upregulation of PD-L1 expression can be blocked by the anti-PD-L1 antibody. Irinotecan 28-38 CD274 antigen Mus musculus 98-103 30140379-8 2018 These results indicate that irinotecan may enhance the effect of T cell activation caused by anti-PD-L1 treatment by reducing Tregs and augmenting MHC class I-mediated tumor antigen presentation, and concurrent upregulation of PD-L1 expression can be blocked by the anti-PD-L1 antibody. Irinotecan 28-38 CD274 antigen Mus musculus 227-232 30140379-8 2018 These results indicate that irinotecan may enhance the effect of T cell activation caused by anti-PD-L1 treatment by reducing Tregs and augmenting MHC class I-mediated tumor antigen presentation, and concurrent upregulation of PD-L1 expression can be blocked by the anti-PD-L1 antibody. Irinotecan 28-38 CD274 antigen Mus musculus 227-232 30131854-8 2018 Moreover, FSTL1 knockdown made CRC cells more susceptible to oxaliplatin and irinotecan-induced death. Irinotecan 77-87 follistatin like 1 Homo sapiens 10-15 29728798-0 2018 UGT1A polymorphisms associated with worse outcome in colorectal cancer patients treated with irinotecan-based chemotherapy. Irinotecan 93-103 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 0-5 29728798-1 2018 PURPOSE: To investigate the association between UDP-glucuronosyltransferase (UGT)1A polymorphisms and irinotecan-treatment efficacy in a Chinese population with metastatic colorectal cancer (mCRC). Irinotecan 102-112 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 77-83 29728798-9 2018 CONCLUSIONS: UGT1A polymorphisms are predictive of survival outcome of irinotecan-treated Chinese mCRC patients. Irinotecan 71-81 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 13-18 29842993-1 2018 INTRODUCTION: The coexpression of pIGF-1R and MMP-7 (double-positive phenotype, DP) correlates with poor overall survival (OS) in KRAS wild-type (WT) (exon 2) metastatic colorectal cancer (mCRC) patients treated with irinotecan-cetuximab in second/third line. Irinotecan 217-227 matrix metallopeptidase 7 Homo sapiens 46-51 30075835-8 2018 Pharmacogenomics guidelines have been published by some international scientific consortia such as the Clinical Pharmacogenomics Implementation Consortium (CPIC) and the Dutch Pharmacogenetics Working Group (DPWG) strongly suggesting a pre-treatment dose adjustment of irinotecan based on UGT1A1*28 genotype and of fluoropyrimidines based on some DPYD genetic variants, to increase treatment safety. Irinotecan 269-279 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 289-295 30075835-8 2018 Pharmacogenomics guidelines have been published by some international scientific consortia such as the Clinical Pharmacogenomics Implementation Consortium (CPIC) and the Dutch Pharmacogenetics Working Group (DPWG) strongly suggesting a pre-treatment dose adjustment of irinotecan based on UGT1A1*28 genotype and of fluoropyrimidines based on some DPYD genetic variants, to increase treatment safety. Irinotecan 269-279 dihydropyrimidine dehydrogenase Homo sapiens 347-351 29977740-6 2018 Results: The GE11/HA/TATp-irinotecan liposomes evidently increased the uptake of irinotecan and showed significant antitumor efficacy in the xenografted A549 cancer cells in nude mice by intravenous administration. Irinotecan 26-36 tyrosine aminotransferase Mus musculus 21-25 29989029-4 2018 When the irinotecan metabolite, SN-38, is conjugated to a humanized anti-TROP-2 antibody (sacituzumab govitecan), it shows potent broad anticancer activity in human cancer xenografts and in patients with advanced triple-negative breast, non-small cell and small-cell lung, as well as urothelial cancers. Irinotecan 9-19 tumor associated calcium signal transducer 2 Homo sapiens 73-79 29989029-4 2018 When the irinotecan metabolite, SN-38, is conjugated to a humanized anti-TROP-2 antibody (sacituzumab govitecan), it shows potent broad anticancer activity in human cancer xenografts and in patients with advanced triple-negative breast, non-small cell and small-cell lung, as well as urothelial cancers. Irinotecan 32-37 tumor associated calcium signal transducer 2 Homo sapiens 73-79 29468758-3 2018 In this study, the inhibitory effects of shikonin on the activity of hCE2 in human liver microsomes are investigated by using fluorescein diacetate (FD), N-(2-butyl-1,3-dioxo-2,3-dihydro-1H-phenalen-6-yl)-2-chloroacetamide (NCEN), and CPT-11 as substrates of hCE2. Irinotecan 235-241 carboxylesterase 2 Homo sapiens 69-73 29468758-4 2018 The results demonstrate that shikonin significantly inhibits the activity of hCE2 when FD and NCEN are used as substrates, whereas the half inhibition concentration value of shikonin increased by 5-30 times when CPT-11 was used as the substrate. Irinotecan 212-218 carboxylesterase 2 Homo sapiens 77-81 29518481-3 2018 The efficacy of SN-38, topotecan, and mitoxantrone (MX) were significantly increased by PD153035, PD153035 significantly reversed ABCG2-mediated MDR by attenuating the efflux activity of this transporter. Irinotecan 16-21 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 130-135 29977740-6 2018 Results: The GE11/HA/TATp-irinotecan liposomes evidently increased the uptake of irinotecan and showed significant antitumor efficacy in the xenografted A549 cancer cells in nude mice by intravenous administration. Irinotecan 81-91 tyrosine aminotransferase Mus musculus 21-25 29930754-1 2018 Nrf2 is a transcription factor that regulates cellular stress response and irinotecan-metabolising pathways. Irinotecan 75-85 nuclear factor, erythroid derived 2, like 2 Mus musculus 0-4 29121274-1 2018 Background: The GLARIUS trial, which investigated the efficacy of bevacizumab (BEV)/irinotecan (IRI) compared with standard temozolomide in the first-line therapy of O6-methylguanine-DNA methyltransferase (MGMT)-nonmethylated glioblastoma, showed that progression-free survival was significantly prolonged by BEV/IRI, while overall survival was similar in both arms. Irinotecan 84-94 O-6-methylguanine-DNA methyltransferase Homo sapiens 166-204 29963241-3 2018 Here we demonstrate that CPT-11 stalls such cells in the G2/M phase of the cell cycle, induces an accumulation of the tumor suppressor p53, the replicative stress/DNA damage marker gammaH2AX, phosphorylation of the checkpoint kinases ATM and ATR, and an ATR-dependent accumulation of the pro-survival molecule survivin. Irinotecan 25-31 tumor protein p53 Homo sapiens 135-138 29963241-3 2018 Here we demonstrate that CPT-11 stalls such cells in the G2/M phase of the cell cycle, induces an accumulation of the tumor suppressor p53, the replicative stress/DNA damage marker gammaH2AX, phosphorylation of the checkpoint kinases ATM and ATR, and an ATR-dependent accumulation of the pro-survival molecule survivin. Irinotecan 25-31 ATM serine/threonine kinase Homo sapiens 234-237 29963241-3 2018 Here we demonstrate that CPT-11 stalls such cells in the G2/M phase of the cell cycle, induces an accumulation of the tumor suppressor p53, the replicative stress/DNA damage marker gammaH2AX, phosphorylation of the checkpoint kinases ATM and ATR, and an ATR-dependent accumulation of the pro-survival molecule survivin. Irinotecan 25-31 ATR serine/threonine kinase Homo sapiens 242-245 29963241-3 2018 Here we demonstrate that CPT-11 stalls such cells in the G2/M phase of the cell cycle, induces an accumulation of the tumor suppressor p53, the replicative stress/DNA damage marker gammaH2AX, phosphorylation of the checkpoint kinases ATM and ATR, and an ATR-dependent accumulation of the pro-survival molecule survivin. Irinotecan 25-31 ATR serine/threonine kinase Homo sapiens 254-257 29738245-8 2018 In an intraperitoneal tumor xenograft model mimicking late-stage metastatic cervical cancer, the LPQDEA/DNA vector with TRAIL suicide gene exerted strong tumor inhibition as effective as paclitaxel, the first-line anticancer drug, but gave much less tumor relapse and much longer survival than the clinical chemotherapy drugs, irinotecan and paclitaxel. Irinotecan 327-337 TNF superfamily member 10 Homo sapiens 120-125 29930754-5 2018 In vitro experiments in CRC cell lines revealed that Nrf2 siRNA and brusatol, which is known to inhibit Nrf2, decreased viability and sensitised cells to irinotecan toxicity. Irinotecan 154-164 nuclear factor, erythroid derived 2, like 2 Mus musculus 53-57 29653076-8 2018 E4bp4-/- mice also showed reduced Ces-mediated metabolism and elevated systemic exposure of CPT-11, a well-known Ces substrate. Irinotecan 92-98 nuclear factor, interleukin 3, regulated Mus musculus 0-5 29688721-0 2018 Nanoencapsulation of Novel Inhibitors of PNKP for Selective Sensitization to Ionizing Radiation and Irinotecan and Induction of Synthetic Lethality. Irinotecan 100-110 polynucleotide kinase 3'-phosphatase Homo sapiens 41-45 29848680-0 2018 SN-38 Acts as a Radiosensitizer for Colorectal Cancer by Inhibiting the Radiation-induced Up-regulation of HIF-1alpha. Irinotecan 0-5 hypoxia inducible factor 1 subunit alpha Homo sapiens 107-117 29848680-2 2018 The aim of the study was to investigate whether SN-38, the active metabolite of irinotecan, acts as a radiosensitizer through inhibition of hypoxia-inducible factor (HIF)-1alpha in the human colorectal cancer (CRC) cells. Irinotecan 48-53 hypoxia inducible factor 1 subunit alpha Homo sapiens 140-177 29848680-2 2018 The aim of the study was to investigate whether SN-38, the active metabolite of irinotecan, acts as a radiosensitizer through inhibition of hypoxia-inducible factor (HIF)-1alpha in the human colorectal cancer (CRC) cells. Irinotecan 80-90 hypoxia inducible factor 1 subunit alpha Homo sapiens 140-177 29848680-3 2018 MATERIALS AND METHODS: HT29 and SW480 cells were cultured with SN-38 (0-4 muM) immediately after irradiation (0-8 Gy). Irinotecan 63-68 latexin Homo sapiens 74-77 29848680-7 2018 RESULTS: Radiation up-regulated HIF-1alpha, and SN-38 inhibited the radiation-induced HIF-1alpha. Irinotecan 48-53 hypoxia inducible factor 1 subunit alpha Homo sapiens 86-96 29848680-8 2018 The combination of radiation and SN-38 inhibited cell proliferation more than radiation alone; treatment with SN-38 after radiation exposure did not increase the number of apoptotic cells, whereas, it enhanced the S and G2/M cell-cycle arrest and decreased the population of cells in G1 Conclusion: SN-38 inhibits the radiation-induced up-regulation of HIF-1alpha and acts as a radiosensitizer by inducing cell-cycle arrest in CRC cells. Irinotecan 33-38 hypoxia inducible factor 1 subunit alpha Homo sapiens 353-363 29848680-8 2018 The combination of radiation and SN-38 inhibited cell proliferation more than radiation alone; treatment with SN-38 after radiation exposure did not increase the number of apoptotic cells, whereas, it enhanced the S and G2/M cell-cycle arrest and decreased the population of cells in G1 Conclusion: SN-38 inhibits the radiation-induced up-regulation of HIF-1alpha and acts as a radiosensitizer by inducing cell-cycle arrest in CRC cells. Irinotecan 110-115 hypoxia inducible factor 1 subunit alpha Homo sapiens 353-363 29848680-8 2018 The combination of radiation and SN-38 inhibited cell proliferation more than radiation alone; treatment with SN-38 after radiation exposure did not increase the number of apoptotic cells, whereas, it enhanced the S and G2/M cell-cycle arrest and decreased the population of cells in G1 Conclusion: SN-38 inhibits the radiation-induced up-regulation of HIF-1alpha and acts as a radiosensitizer by inducing cell-cycle arrest in CRC cells. Irinotecan 110-115 hypoxia inducible factor 1 subunit alpha Homo sapiens 353-363 29504153-0 2018 Relevance of CYP3A4*20, UGT1A1*37 and UGT1A1*28 variants in irinotecan-induced severe toxicity. Irinotecan 60-70 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 13-19 29653076-2 2018 Here, we investigate a potential role of E4 promoter-binding protein 4 (E4bp4) in regulation of Ces and CPT-11 (irinotecan, a first-line drug for treating colorectal cancer) pharmacokinetics in mice. Irinotecan 104-110 nuclear factor, interleukin 3, regulated Mus musculus 41-70 29653076-2 2018 Here, we investigate a potential role of E4 promoter-binding protein 4 (E4bp4) in regulation of Ces and CPT-11 (irinotecan, a first-line drug for treating colorectal cancer) pharmacokinetics in mice. Irinotecan 104-110 nuclear factor, interleukin 3, regulated Mus musculus 72-77 29653076-2 2018 Here, we investigate a potential role of E4 promoter-binding protein 4 (E4bp4) in regulation of Ces and CPT-11 (irinotecan, a first-line drug for treating colorectal cancer) pharmacokinetics in mice. Irinotecan 112-122 nuclear factor, interleukin 3, regulated Mus musculus 41-70 29653076-2 2018 Here, we investigate a potential role of E4 promoter-binding protein 4 (E4bp4) in regulation of Ces and CPT-11 (irinotecan, a first-line drug for treating colorectal cancer) pharmacokinetics in mice. Irinotecan 112-122 nuclear factor, interleukin 3, regulated Mus musculus 72-77 29504153-0 2018 Relevance of CYP3A4*20, UGT1A1*37 and UGT1A1*28 variants in irinotecan-induced severe toxicity. Irinotecan 60-70 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 24-30 29504153-0 2018 Relevance of CYP3A4*20, UGT1A1*37 and UGT1A1*28 variants in irinotecan-induced severe toxicity. Irinotecan 60-70 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 38-44 29873288-1 2018 BACKGROUND: Irinotecan (IRN) (CPT-11) is a camptothecin derivative with low oral bioavailability due to active efflux by intestinal P-glycoprotein (p-gp) receptors. Irinotecan 12-22 ATP binding cassette subfamily B member 1 Homo sapiens 132-146 29504153-1 2018 Severe irinotecan-induced toxicity is associated with UGT1A1 polymorphisms. Irinotecan 7-17 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 54-60 29504153-3 2018 As CYP3A4 is also an irinotecan-metabolizing enzyme, our study aimed to elucidate the influence of the CYP3A4*20 loss-of-function allele in the toxicity profile of these patients. Irinotecan 21-31 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 3-9 29504153-3 2018 As CYP3A4 is also an irinotecan-metabolizing enzyme, our study aimed to elucidate the influence of the CYP3A4*20 loss-of-function allele in the toxicity profile of these patients. Irinotecan 21-31 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 103-109 29873288-1 2018 BACKGROUND: Irinotecan (IRN) (CPT-11) is a camptothecin derivative with low oral bioavailability due to active efflux by intestinal P-glycoprotein (p-gp) receptors. Irinotecan 24-27 ATP binding cassette subfamily B member 1 Homo sapiens 132-146 29873288-1 2018 BACKGROUND: Irinotecan (IRN) (CPT-11) is a camptothecin derivative with low oral bioavailability due to active efflux by intestinal P-glycoprotein (p-gp) receptors. Irinotecan 30-36 ATP binding cassette subfamily B member 1 Homo sapiens 132-146 29706892-6 2018 VDR rs11574077 polymorphism was demonstrated to affect both irinotecan biliary index (BI) and glucuronidation ratio (GR) by a pharmacokinetic analysis. Irinotecan 60-70 vitamin D receptor Homo sapiens 0-3 29899822-6 2018 Furthermore, treatment of aRMS cells with clinically relevant CPT derivative irinotecan restores MyoD function, and myogenic differentiation in vitro and in a xenograft model. Irinotecan 77-87 myogenic differentiation 1 Homo sapiens 97-101 29861877-5 2018 Contrasting results, however, have been reported on the capability of variants of other genes as MTHFR, TYMS, ERCC1, XRCC1, GSTP1, CYP3A4/3A5 and ABCB1, in predicting either therapy efficacy or toxicity in patients undergoing treatment with pyrimidine antimetabolites, platinum derivatives, irinotecan and taxanes. Irinotecan 291-301 methylenetetrahydrofolate reductase Homo sapiens 97-102 29861877-5 2018 Contrasting results, however, have been reported on the capability of variants of other genes as MTHFR, TYMS, ERCC1, XRCC1, GSTP1, CYP3A4/3A5 and ABCB1, in predicting either therapy efficacy or toxicity in patients undergoing treatment with pyrimidine antimetabolites, platinum derivatives, irinotecan and taxanes. Irinotecan 291-301 thymidylate synthetase Homo sapiens 104-108 29861877-5 2018 Contrasting results, however, have been reported on the capability of variants of other genes as MTHFR, TYMS, ERCC1, XRCC1, GSTP1, CYP3A4/3A5 and ABCB1, in predicting either therapy efficacy or toxicity in patients undergoing treatment with pyrimidine antimetabolites, platinum derivatives, irinotecan and taxanes. Irinotecan 291-301 ERCC excision repair 1, endonuclease non-catalytic subunit Homo sapiens 110-115 29861877-5 2018 Contrasting results, however, have been reported on the capability of variants of other genes as MTHFR, TYMS, ERCC1, XRCC1, GSTP1, CYP3A4/3A5 and ABCB1, in predicting either therapy efficacy or toxicity in patients undergoing treatment with pyrimidine antimetabolites, platinum derivatives, irinotecan and taxanes. Irinotecan 291-301 ATP binding cassette subfamily B member 1 Homo sapiens 146-151 29649683-9 2018 This effect contradicted irinotecan mediated UGT inhibition. Irinotecan 25-35 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 45-48 29861877-4 2018 Among the examined germline polymorphic variants, several SNPs of dihydropyrimidine dehydrogenase (DPYD) and uridine diphosphate glucuronosyltransferases (UGT) have shown a robust role as predictors of toxicity following fluoropyrimidine- and/or irinotecan-based treatments respectively, and a few guidelines are mandatory in their detection before therapy initiation. Irinotecan 246-256 dihydropyrimidine dehydrogenase Homo sapiens 66-97 29861877-4 2018 Among the examined germline polymorphic variants, several SNPs of dihydropyrimidine dehydrogenase (DPYD) and uridine diphosphate glucuronosyltransferases (UGT) have shown a robust role as predictors of toxicity following fluoropyrimidine- and/or irinotecan-based treatments respectively, and a few guidelines are mandatory in their detection before therapy initiation. Irinotecan 246-256 dihydropyrimidine dehydrogenase Homo sapiens 99-103 29861877-4 2018 Among the examined germline polymorphic variants, several SNPs of dihydropyrimidine dehydrogenase (DPYD) and uridine diphosphate glucuronosyltransferases (UGT) have shown a robust role as predictors of toxicity following fluoropyrimidine- and/or irinotecan-based treatments respectively, and a few guidelines are mandatory in their detection before therapy initiation. Irinotecan 246-256 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 109-153 29861877-4 2018 Among the examined germline polymorphic variants, several SNPs of dihydropyrimidine dehydrogenase (DPYD) and uridine diphosphate glucuronosyltransferases (UGT) have shown a robust role as predictors of toxicity following fluoropyrimidine- and/or irinotecan-based treatments respectively, and a few guidelines are mandatory in their detection before therapy initiation. Irinotecan 246-256 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 155-158 29760556-4 2018 Patients treated by anti-epidermal growth factor receptor (EGFR) antibodies with irinotecan were included in the control arm. Irinotecan 81-91 epidermal growth factor receptor Homo sapiens 20-57 29760556-4 2018 Patients treated by anti-epidermal growth factor receptor (EGFR) antibodies with irinotecan were included in the control arm. Irinotecan 81-91 epidermal growth factor receptor Homo sapiens 59-63 29760556-12 2018 The present study found that rechallenge of oxaliplatin-containing regimens produced equivalent tumor control and survival benefit to that of anti-EGFR antibodies with irinotecan in mCRC. Irinotecan 168-178 epidermal growth factor receptor Homo sapiens 147-151 29713498-1 2018 Background: In the RAISE trial, ramucirumab+leucovorin/fluorouracil/irinotecan (FOLFIRI) improved the median overall survival (mOS) of patients with previously treated metastatic colorectal cancer versus patients treated with placebo+FOLFIRI but had a higher incidence of neutropaenia, leading to more chemotherapy dose modifications and discontinuations. Irinotecan 68-78 Moloney sarcoma oncogene Mus musculus 127-130 29076612-0 2018 New Insights into SN-38 Glucuronidation: Evidence for the Important Role of UDP Glucuronosyltransferase 1A9. Irinotecan 18-23 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 76-107 29420338-9 2018 After curcumin treatment, drug-resistant cell proliferation was significantly inhibited; in the curcumin+irinotecan treatment group, E-cadherin expression was upregulated, whereas vimentin and N-cadherin expressions were downregulated. Irinotecan 105-115 cadherin 1 Homo sapiens 133-143 29420338-9 2018 After curcumin treatment, drug-resistant cell proliferation was significantly inhibited; in the curcumin+irinotecan treatment group, E-cadherin expression was upregulated, whereas vimentin and N-cadherin expressions were downregulated. Irinotecan 105-115 cadherin 2 Homo sapiens 193-203 29599307-10 2018 Irinotecan, 5-FU and 50 mg/kg EEG significantly decreased the protein expression of beta-catenin and MMP-7. Irinotecan 0-10 catenin beta 1 Homo sapiens 84-96 29599307-10 2018 Irinotecan, 5-FU and 50 mg/kg EEG significantly decreased the protein expression of beta-catenin and MMP-7. Irinotecan 0-10 matrix metallopeptidase 7 Homo sapiens 101-106 29599307-11 2018 Cyclin D1 expression was decreased and E-cadherin expression was increased by irinotecan, 5-FU and all three doses of EEG. Irinotecan 78-88 cadherin 1 Homo sapiens 39-49 28520360-0 2012 Irinotecan Therapy and UGT1A1 Genotype Irinotecan is a topoisomerase inhibitor that is widely used in the treatment of cancer. Irinotecan 39-49 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 23-29 28520360-3 2012 Irinotecan is metabolized and inactivated by an UDP-glucuronosyltransferase enzyme encoded by the gene UGT1A1. Irinotecan 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 103-109 28520360-5 2012 Variants of this gene, such as UGT1A1*28, are associated with reduced enzyme activity and an increased risk of irinotecan toxicity. Irinotecan 111-121 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 31-37 28520360-6 2012 Approximately 10% of North Americans are homozygous for the UGT1A1*28 allele and are more likely to develop neutropenia following irinotecan therapy (3). Irinotecan 130-140 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 60-66 28520360-7 2012 The FDA-approved drug label for irinotecan states that "when administered as a single-agent, a reduction in the starting dose by at least one level of irinotecan hydrochloride injection should be considered for patients known to be homozygous for the UGT1A1*28 allele. Irinotecan 32-42 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 251-257 28520360-7 2012 The FDA-approved drug label for irinotecan states that "when administered as a single-agent, a reduction in the starting dose by at least one level of irinotecan hydrochloride injection should be considered for patients known to be homozygous for the UGT1A1*28 allele. Irinotecan 151-175 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 251-257 28520360-9 2012 A guideline from the Dutch Pharmacogenetics Working Group (KNMP) mentions "although results are not consistent, there is sufficient evidence that a reduction in the initial dose by 30% is required for regimens containing >250 mg/m(2) of irinotecan prescribed to homozygous carriers of the UGT1A1*28 allele. Irinotecan 240-250 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 292-298 29076612-6 2018 For UGT1A9 and HLM, BSA can significantly accelerate SN-38 glucuronidation activities and similar effects are further observed on kinetic patterns. Irinotecan 53-58 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 4-10 29076612-9 2018 To get the true contribution of UGT1A9 in SN-38 glucuronidation, a relative activity factor (RAF) approach was additionally used. Irinotecan 42-47 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 32-38 29076612-12 2018 Results together conclude that UGT1A9 serves as an additional important contributor to hepatic SN-38 glucuronidation. Irinotecan 95-100 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 31-37 29421708-2 2018 Among all identified hCEs, human carboxylesterase 2 (hCE2) plays crucial roles in the metabolic activation of ester drugs including irinotecan and flutamide. Irinotecan 132-142 carboxylesterase 2 Homo sapiens 33-51 29421708-2 2018 Among all identified hCEs, human carboxylesterase 2 (hCE2) plays crucial roles in the metabolic activation of ester drugs including irinotecan and flutamide. Irinotecan 132-142 carboxylesterase 2 Homo sapiens 53-57 29541236-3 2018 DAC potentiated the antitumor activity of CPT-11 additively, and that of SN-38 synergistically, as measured by colony formation in the human colorectal cancer HCT116 cell line. Irinotecan 42-48 arylacetamide deacetylase Homo sapiens 0-3 28053052-12 2018 CONCLUSIONS: Irinotecan induces hepatic steatosis via autophagy impairment and inflammation via ERK activation. Irinotecan 13-23 mitogen-activated protein kinase 1 Homo sapiens 96-99 29075780-10 2018 Higher HER3 was predictive, being associated with prolonged PFS on irinotecan plus panitumumab (IrPan) (HR, 0.71; 95% CI, 0.61-0.82; P < .001), but not irinotecan (HR, 0.96; 95% CI, 0.82-1.13; P = .65) in patients with RAS wt, with significant interaction between biomarker and treatment (P = .001). Irinotecan 67-77 erb-b2 receptor tyrosine kinase 3 Homo sapiens 7-11 29075780-10 2018 Higher HER3 was predictive, being associated with prolonged PFS on irinotecan plus panitumumab (IrPan) (HR, 0.71; 95% CI, 0.61-0.82; P < .001), but not irinotecan (HR, 0.96; 95% CI, 0.82-1.13; P = .65) in patients with RAS wt, with significant interaction between biomarker and treatment (P = .001). Irinotecan 155-165 erb-b2 receptor tyrosine kinase 3 Homo sapiens 7-11 29075780-12 2018 In an exploratory binary model, dividing the population at the 66th percentile, HER3 was predictive of panitumumab benefit: in patients with high HER3 expression, median PFS was 8.2 months (IrPan) vs 4.4 months (irinotecan) (HR, 0.33; 95% CI, 0.19-0.58; P < .001). Irinotecan 212-222 erb-b2 receptor tyrosine kinase 3 Homo sapiens 80-84 29541236-5 2018 Anti-apoptotic B-cell lymphoma-2 (Bcl-2) protein expression was reduced to 50-67% of the control following a single treatment with CPT-11, SN-38, or DAC, and was markedly reduced to 7-8% following the combination of CPT-11/SN-38 with DAC. Irinotecan 131-137 arylacetamide deacetylase Homo sapiens 234-237 29541236-5 2018 Anti-apoptotic B-cell lymphoma-2 (Bcl-2) protein expression was reduced to 50-67% of the control following a single treatment with CPT-11, SN-38, or DAC, and was markedly reduced to 7-8% following the combination of CPT-11/SN-38 with DAC. Irinotecan 139-144 BCL2 apoptosis regulator Homo sapiens 34-39 29541236-5 2018 Anti-apoptotic B-cell lymphoma-2 (Bcl-2) protein expression was reduced to 50-67% of the control following a single treatment with CPT-11, SN-38, or DAC, and was markedly reduced to 7-8% following the combination of CPT-11/SN-38 with DAC. Irinotecan 216-222 BCL2 apoptosis regulator Homo sapiens 34-39 29541236-5 2018 Anti-apoptotic B-cell lymphoma-2 (Bcl-2) protein expression was reduced to 50-67% of the control following a single treatment with CPT-11, SN-38, or DAC, and was markedly reduced to 7-8% following the combination of CPT-11/SN-38 with DAC. Irinotecan 223-228 BCL2 apoptosis regulator Homo sapiens 34-39 29541236-7 2018 Wilms" tumor protein (WT1), which has been shown to be a positive regulator of Bcl-2 in HCT116 cells through WT1-kncokdown experiments, was downregulated in HCT116 and HT29 cells when treated with CPT-11/SN-38 combined with DAC, with decreases greater than any single administration of CPT-11, SN-38, or DAC. Irinotecan 197-203 WT1 transcription factor Homo sapiens 0-20 29541236-7 2018 Wilms" tumor protein (WT1), which has been shown to be a positive regulator of Bcl-2 in HCT116 cells through WT1-kncokdown experiments, was downregulated in HCT116 and HT29 cells when treated with CPT-11/SN-38 combined with DAC, with decreases greater than any single administration of CPT-11, SN-38, or DAC. Irinotecan 197-203 WT1 transcription factor Homo sapiens 22-25 29541236-7 2018 Wilms" tumor protein (WT1), which has been shown to be a positive regulator of Bcl-2 in HCT116 cells through WT1-kncokdown experiments, was downregulated in HCT116 and HT29 cells when treated with CPT-11/SN-38 combined with DAC, with decreases greater than any single administration of CPT-11, SN-38, or DAC. Irinotecan 197-203 BCL2 apoptosis regulator Homo sapiens 79-84 29541236-7 2018 Wilms" tumor protein (WT1), which has been shown to be a positive regulator of Bcl-2 in HCT116 cells through WT1-kncokdown experiments, was downregulated in HCT116 and HT29 cells when treated with CPT-11/SN-38 combined with DAC, with decreases greater than any single administration of CPT-11, SN-38, or DAC. Irinotecan 204-209 WT1 transcription factor Homo sapiens 0-20 29541236-7 2018 Wilms" tumor protein (WT1), which has been shown to be a positive regulator of Bcl-2 in HCT116 cells through WT1-kncokdown experiments, was downregulated in HCT116 and HT29 cells when treated with CPT-11/SN-38 combined with DAC, with decreases greater than any single administration of CPT-11, SN-38, or DAC. Irinotecan 204-209 WT1 transcription factor Homo sapiens 22-25 29541236-7 2018 Wilms" tumor protein (WT1), which has been shown to be a positive regulator of Bcl-2 in HCT116 cells through WT1-kncokdown experiments, was downregulated in HCT116 and HT29 cells when treated with CPT-11/SN-38 combined with DAC, with decreases greater than any single administration of CPT-11, SN-38, or DAC. Irinotecan 204-209 BCL2 apoptosis regulator Homo sapiens 79-84 29541236-7 2018 Wilms" tumor protein (WT1), which has been shown to be a positive regulator of Bcl-2 in HCT116 cells through WT1-kncokdown experiments, was downregulated in HCT116 and HT29 cells when treated with CPT-11/SN-38 combined with DAC, with decreases greater than any single administration of CPT-11, SN-38, or DAC. Irinotecan 286-292 WT1 transcription factor Homo sapiens 0-20 29541236-7 2018 Wilms" tumor protein (WT1), which has been shown to be a positive regulator of Bcl-2 in HCT116 cells through WT1-kncokdown experiments, was downregulated in HCT116 and HT29 cells when treated with CPT-11/SN-38 combined with DAC, with decreases greater than any single administration of CPT-11, SN-38, or DAC. Irinotecan 286-292 WT1 transcription factor Homo sapiens 22-25 28053052-0 2018 ERK activation and autophagy impairment are central mediators of irinotecan-induced steatohepatitis. Irinotecan 65-75 mitogen-activated protein kinase 1 Homo sapiens 0-3 28053052-6 2018 ERK inhibition prevented irinotecan-induced pro-inflammatory gene expression but had only a slight effect on lipid accumulation. Irinotecan 25-35 mitogen-activated protein kinase 1 Homo sapiens 0-3 28053052-9 2018 Also in mice, irinotecan treatment induced hepatic ACOX1 expression, ERK phosphorylation and inflammation, as well as impairment of autophagy and significant steatosis. Irinotecan 14-24 acyl-Coenzyme A oxidase 1, palmitoyl Mus musculus 51-56 28053052-9 2018 Also in mice, irinotecan treatment induced hepatic ACOX1 expression, ERK phosphorylation and inflammation, as well as impairment of autophagy and significant steatosis. Irinotecan 14-24 mitogen-activated protein kinase 1 Mus musculus 69-72 28053052-10 2018 Furthermore, irinotecan-treated patients revealed higher hepatic ERK activity, expression of pro-inflammatory genes and markers indicative for a shift to peroxisomal fatty acid oxidation and an impaired autophagic flux. Irinotecan 13-23 mitogen-activated protein kinase 1 Homo sapiens 65-68 28053052-11 2018 Pretreatment with the multityrosine kinase inhibitor sorafenib did not affect autophagy impairment and steatosis but significantly reduced ERK phosphorylation and inflammatory response in irinotecan-treated hepatocytes and murine livers. Irinotecan 188-198 mitogen-activated protein kinase 1 Mus musculus 139-142 29541236-7 2018 Wilms" tumor protein (WT1), which has been shown to be a positive regulator of Bcl-2 in HCT116 cells through WT1-kncokdown experiments, was downregulated in HCT116 and HT29 cells when treated with CPT-11/SN-38 combined with DAC, with decreases greater than any single administration of CPT-11, SN-38, or DAC. Irinotecan 286-292 BCL2 apoptosis regulator Homo sapiens 79-84 29541236-7 2018 Wilms" tumor protein (WT1), which has been shown to be a positive regulator of Bcl-2 in HCT116 cells through WT1-kncokdown experiments, was downregulated in HCT116 and HT29 cells when treated with CPT-11/SN-38 combined with DAC, with decreases greater than any single administration of CPT-11, SN-38, or DAC. Irinotecan 294-299 WT1 transcription factor Homo sapiens 0-20 29541236-7 2018 Wilms" tumor protein (WT1), which has been shown to be a positive regulator of Bcl-2 in HCT116 cells through WT1-kncokdown experiments, was downregulated in HCT116 and HT29 cells when treated with CPT-11/SN-38 combined with DAC, with decreases greater than any single administration of CPT-11, SN-38, or DAC. Irinotecan 294-299 WT1 transcription factor Homo sapiens 22-25 29541236-5 2018 Anti-apoptotic B-cell lymphoma-2 (Bcl-2) protein expression was reduced to 50-67% of the control following a single treatment with CPT-11, SN-38, or DAC, and was markedly reduced to 7-8% following the combination of CPT-11/SN-38 with DAC. Irinotecan 131-137 BCL2 apoptosis regulator Homo sapiens 34-39 29541236-7 2018 Wilms" tumor protein (WT1), which has been shown to be a positive regulator of Bcl-2 in HCT116 cells through WT1-kncokdown experiments, was downregulated in HCT116 and HT29 cells when treated with CPT-11/SN-38 combined with DAC, with decreases greater than any single administration of CPT-11, SN-38, or DAC. Irinotecan 294-299 BCL2 apoptosis regulator Homo sapiens 79-84 29541236-8 2018 The extent of CPT-11/SN-38 potentiation by DAC may depend on Bcl-2 expression levels in human colorectal cancer cells. Irinotecan 14-20 arylacetamide deacetylase Homo sapiens 43-46 29541236-8 2018 The extent of CPT-11/SN-38 potentiation by DAC may depend on Bcl-2 expression levels in human colorectal cancer cells. Irinotecan 14-20 BCL2 apoptosis regulator Homo sapiens 61-66 29541236-8 2018 The extent of CPT-11/SN-38 potentiation by DAC may depend on Bcl-2 expression levels in human colorectal cancer cells. Irinotecan 21-26 arylacetamide deacetylase Homo sapiens 43-46 29541236-8 2018 The extent of CPT-11/SN-38 potentiation by DAC may depend on Bcl-2 expression levels in human colorectal cancer cells. Irinotecan 21-26 BCL2 apoptosis regulator Homo sapiens 61-66 29261002-0 2018 Expression of Topoisomerase 1 and carboxylesterase 2 correlates with irinotecan treatment response in metastatic colorectal cancer. Irinotecan 69-79 carboxylesterase 2 Homo sapiens 34-52 29261002-13 2018 In summary, our findings suggest that TOPO-1 and CES-2 may play important roles irinotecan sensitivity in mCRC patients. Irinotecan 80-90 carboxylesterase 2 Homo sapiens 49-54 29261002-14 2018 Evaluation of expression of TOPO-1 and CES-2 may provide preliminary clinical evidence for the management of irinotecan-based therapy in mCRC patients. Irinotecan 109-119 carboxylesterase 2 Homo sapiens 39-44 31352923-3 2018 In this study, to improve water dispersity and the anti-tumor efficiency of SN-38, a prodrug, SN-38-BOC, that could efficiently transform to active SN-38 in the acidic tumor microenvironment was synthesized and encapsulated into MPEG-P(CL-ran-TMC) micelles (SN-38-BOC micelles). Irinotecan 94-99 BOC cell adhesion associated, oncogene regulated Homo sapiens 100-103 29228087-1 2018 Background: The phase III RAISE trial (NCT01183780) demonstrated that the vascular endothelial growth factor (VEGF) receptor (VEGFR)-2 binding monoclonal antibody ramucirumab plus 5-fluororuracil, leucovorin, and irinotecan (FOLFIRI) significantly improved overall survival (OS) and progression-free survival (PFS) compared with placebo + FOLFIRI as second-line metastatic colorectal cancer (mCRC) treatment. Irinotecan 213-223 kinase insert domain receptor Homo sapiens 126-131 29246888-5 2018 The EC50 of 7-ethyl-10-hydroxycamptothecin (SN-38), topotecan, and mitoxantrone decreased in the presence of a BCRP inhibitor in PANC-1 and AsPC-1 cells, which exhibit high BCRP expression. Irinotecan 12-42 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 111-115 29246888-5 2018 The EC50 of 7-ethyl-10-hydroxycamptothecin (SN-38), topotecan, and mitoxantrone decreased in the presence of a BCRP inhibitor in PANC-1 and AsPC-1 cells, which exhibit high BCRP expression. Irinotecan 12-42 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 173-177 29246888-5 2018 The EC50 of 7-ethyl-10-hydroxycamptothecin (SN-38), topotecan, and mitoxantrone decreased in the presence of a BCRP inhibitor in PANC-1 and AsPC-1 cells, which exhibit high BCRP expression. Irinotecan 44-49 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 111-115 29519997-0 2018 Overexpression of Nitrogen Permease Regulator Like-2 (NPRL2) Enhances Sensitivity to Irinotecan (CPT-11) in Colon Cancer Cells by Activating the DNA Damage Checkpoint Pathway. Irinotecan 85-95 NPR2 like, GATOR1 complex subunit Homo sapiens 18-52 29519997-0 2018 Overexpression of Nitrogen Permease Regulator Like-2 (NPRL2) Enhances Sensitivity to Irinotecan (CPT-11) in Colon Cancer Cells by Activating the DNA Damage Checkpoint Pathway. Irinotecan 85-95 NPR2 like, GATOR1 complex subunit Homo sapiens 54-59 29519997-0 2018 Overexpression of Nitrogen Permease Regulator Like-2 (NPRL2) Enhances Sensitivity to Irinotecan (CPT-11) in Colon Cancer Cells by Activating the DNA Damage Checkpoint Pathway. Irinotecan 97-103 NPR2 like, GATOR1 complex subunit Homo sapiens 18-52 29519997-0 2018 Overexpression of Nitrogen Permease Regulator Like-2 (NPRL2) Enhances Sensitivity to Irinotecan (CPT-11) in Colon Cancer Cells by Activating the DNA Damage Checkpoint Pathway. Irinotecan 97-103 NPR2 like, GATOR1 complex subunit Homo sapiens 54-59 29519997-3 2018 The aim of this study was to investigate the role of NPRL2 in improving sensitivity to CPT-11 in colon cancer cells. Irinotecan 87-93 NPR2 like, GATOR1 complex subunit Homo sapiens 53-58 29519997-10 2018 RESULTS A CCK8 assay revealed that the overexpression of NPRL2 improved the sensitivity of CPT-11 in HCT116 cells (P<0.05). Irinotecan 91-97 NPR2 like, GATOR1 complex subunit Homo sapiens 57-62 29519997-11 2018 Functionally, NPRL2 overexpression elevated the sensitivity of CPT-11 by preventing colon cancer cell proliferation, cell movement, and invasion, and promoting cell apoptosis and G2/M cell cycle arrest. Irinotecan 63-69 NPR2 like, GATOR1 complex subunit Homo sapiens 14-19 29519997-12 2018 Mechanistically, NPRL2 overexpression enhanced CPT-11 sensitivity by activating the DNA damage checkpoint pathway. Irinotecan 47-53 NPR2 like, GATOR1 complex subunit Homo sapiens 17-22 29169974-0 2018 A Prospective, Multicenter Phase II Study of the Efficacy and Feasibility of 15-minute Panitumumab Infusion Plus Irinotecan for Oxaliplatin- and Irinotecan-refractory, KRAS Wild-type Metastatic Colorectal Cancer (Short Infusion of Panitumumab Trial). Irinotecan 113-123 KRAS proto-oncogene, GTPase Homo sapiens 168-172 29328485-12 2018 hnRNP A2/B1 knock-down significantly induced the suppression of proliferation, migration, invasion and also enhancement of apoptosis and reduced the IC50 of lobaplatin and irinotecan. Irinotecan 172-182 heterogeneous nuclear ribonucleoprotein A2/B1 Homo sapiens 0-11 29328485-17 2018 In addition, after silencing hnRNP A2/B1 can increase the sensitivity of cervical cancer cells to lobaplatin and irinotecan. Irinotecan 113-123 heterogeneous nuclear ribonucleoprotein A2/B1 Homo sapiens 29-40 29963254-3 2018 In the animal model for irinotecan (CPT-11) induced CID, Hst could selectively inhibit intestinal carboxylesterase (CES2) and thus reduce the local conversion of CPT-11 to cytotoxic SN-38 which causes intestinal toxicity. Irinotecan 24-34 carboxylesterase 2 Homo sapiens 116-120 29963254-3 2018 In the animal model for irinotecan (CPT-11) induced CID, Hst could selectively inhibit intestinal carboxylesterase (CES2) and thus reduce the local conversion of CPT-11 to cytotoxic SN-38 which causes intestinal toxicity. Irinotecan 36-42 carboxylesterase 2 Homo sapiens 116-120 29963254-6 2018 Moreover, Hst was found to work synergistically with CPT-11 in tumor inhibition by suppressing the tumor"s STAT3 activity and recruiting tumoricidal macrophages into the tumor microenvironment. Irinotecan 53-59 signal transducer and activator of transcription 3 Homo sapiens 107-112 31352923-3 2018 In this study, to improve water dispersity and the anti-tumor efficiency of SN-38, a prodrug, SN-38-BOC, that could efficiently transform to active SN-38 in the acidic tumor microenvironment was synthesized and encapsulated into MPEG-P(CL-ran-TMC) micelles (SN-38-BOC micelles). Irinotecan 76-81 BOC cell adhesion associated, oncogene regulated Homo sapiens 100-103 31352923-3 2018 In this study, to improve water dispersity and the anti-tumor efficiency of SN-38, a prodrug, SN-38-BOC, that could efficiently transform to active SN-38 in the acidic tumor microenvironment was synthesized and encapsulated into MPEG-P(CL-ran-TMC) micelles (SN-38-BOC micelles). Irinotecan 76-81 BOC cell adhesion associated, oncogene regulated Homo sapiens 264-267 31352923-3 2018 In this study, to improve water dispersity and the anti-tumor efficiency of SN-38, a prodrug, SN-38-BOC, that could efficiently transform to active SN-38 in the acidic tumor microenvironment was synthesized and encapsulated into MPEG-P(CL-ran-TMC) micelles (SN-38-BOC micelles). Irinotecan 94-99 BOC cell adhesion associated, oncogene regulated Homo sapiens 264-267 31352923-3 2018 In this study, to improve water dispersity and the anti-tumor efficiency of SN-38, a prodrug, SN-38-BOC, that could efficiently transform to active SN-38 in the acidic tumor microenvironment was synthesized and encapsulated into MPEG-P(CL-ran-TMC) micelles (SN-38-BOC micelles). Irinotecan 94-99 BOC cell adhesion associated, oncogene regulated Homo sapiens 100-103 31352923-3 2018 In this study, to improve water dispersity and the anti-tumor efficiency of SN-38, a prodrug, SN-38-BOC, that could efficiently transform to active SN-38 in the acidic tumor microenvironment was synthesized and encapsulated into MPEG-P(CL-ran-TMC) micelles (SN-38-BOC micelles). Irinotecan 94-99 BOC cell adhesion associated, oncogene regulated Homo sapiens 264-267 31352923-4 2018 SN-38-BOC micelles could accumulate in tumors due to the EPR effect and exhibit a sustained release behavior, which was rapidly transformed to active SN-38 by the acidic environment of tumor tissues (pH 5.5-6.8), thus achieving efficient anti-tumor activity. Irinotecan 0-5 BOC cell adhesion associated, oncogene regulated Homo sapiens 6-9 31352923-4 2018 SN-38-BOC micelles could accumulate in tumors due to the EPR effect and exhibit a sustained release behavior, which was rapidly transformed to active SN-38 by the acidic environment of tumor tissues (pH 5.5-6.8), thus achieving efficient anti-tumor activity. Irinotecan 150-155 BOC cell adhesion associated, oncogene regulated Homo sapiens 6-9 31352923-5 2018 Compared with the free SN-38-BOC group, enhanced cytotoxicity and apoptotic induction were obtained from the SN-38-BOC micelle-treated group in both HCT116 and CT26 cells. Irinotecan 23-28 BOC cell adhesion associated, oncogene regulated Homo sapiens 29-32 29167313-2 2018 We show that the antibiotic minocycline, by its ability to minimize DNA repair via reduced expression of tyrosyl-DNA phosphodiesterase-1 (Tdp1), removes a key process attenuating the efficacy of irinotecan, a frequently used chemotherapeutic against metastatic disease. Irinotecan 195-205 tyrosyl-DNA phosphodiesterase 1 Mus musculus 105-136 29167313-2 2018 We show that the antibiotic minocycline, by its ability to minimize DNA repair via reduced expression of tyrosyl-DNA phosphodiesterase-1 (Tdp1), removes a key process attenuating the efficacy of irinotecan, a frequently used chemotherapeutic against metastatic disease. Irinotecan 195-205 tyrosyl-DNA phosphodiesterase 1 Mus musculus 138-142 29167313-3 2018 Moreover, minocycline and irinotecan cooperatively mitigate each other"s undesired cytokine inductions of VEGF and IL8, respectively, thereby reinforcing the benefits of each modality. Irinotecan 26-36 vascular endothelial growth factor A Mus musculus 106-110 29167313-3 2018 Moreover, minocycline and irinotecan cooperatively mitigate each other"s undesired cytokine inductions of VEGF and IL8, respectively, thereby reinforcing the benefits of each modality. Irinotecan 26-36 chemokine (C-X-C motif) ligand 15 Mus musculus 115-118 29257266-0 2018 Wee1 inhibition can suppress tumor proliferation and sensitize p53 mutant colonic cancer cells to the anticancer effect of irinotecan. Irinotecan 123-133 WEE1 G2 checkpoint kinase Homo sapiens 0-4 29257266-0 2018 Wee1 inhibition can suppress tumor proliferation and sensitize p53 mutant colonic cancer cells to the anticancer effect of irinotecan. Irinotecan 123-133 tumor protein p53 Homo sapiens 63-66 30166915-0 2018 Simvastatin enhances irinotecan-induced apoptosis in prostate cancer via inhibition of MCL-1. Irinotecan 21-31 MCL1 apoptosis regulator, BCL2 family member Homo sapiens 87-92 30166915-6 2018 PC3 cells were treated with simvastatin, irinotecan or combination. Irinotecan 41-51 proprotein convertase subtilisin/kexin type 1 Homo sapiens 0-3 30166915-11 2018 Combined treatment of simvastatin with irinotecan exhibited a significant inhibition of PC3 cell growth compared to each treatment alone. Irinotecan 39-49 proprotein convertase subtilisin/kexin type 1 Homo sapiens 88-91 30166915-12 2018 Flow cytometry analysis showed that PC3 cell treatment with simvastatin and irinotecan combination demonstrated a remarkable increase in the percentage of apoptotic cells and those accumulated at G0/G1 phase when compared to each treatment alone. Irinotecan 76-86 proprotein convertase subtilisin/kexin type 1 Homo sapiens 36-39 29651325-1 2018 Irinotecan (CPT-11) is an anticancer prodrug that is activated by the carboxylesterase CES2 and has been approved for the treatment of many types of solid tumors, including colorectal cancer. Irinotecan 0-10 carboxylesterase 2 Homo sapiens 70-86 29124328-1 2018 PURPOSE: We aimed to retrospectively evaluate the efficacy and toxicity of an irinotecan hydrochloride (CPT) and nedaplatin (N) combination therapy for recurrent and refractory endometrial carcinoma, administered based on UGT1A1 genotype. Irinotecan 78-102 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 222-228 29124328-9 2018 Patients with a wild-type UGT1A1 status received higher doses of CPT-11 (p = 0.048) and had similar RR and CBR compared to those with a UGT1A1*6 and *28 status. Irinotecan 65-71 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 26-32 29486476-8 2018 RESULTS: Voruciclib significantly potentiated the effect of paclitaxel and doxorubicin in ABCB1-overexpressing cells, as well as mitoxantrone and SN-38 in ABCG2-overexpressing cells. Irinotecan 146-151 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 155-160 29651325-1 2018 Irinotecan (CPT-11) is an anticancer prodrug that is activated by the carboxylesterase CES2 and has been approved for the treatment of many types of solid tumors, including colorectal cancer. Irinotecan 0-10 carboxylesterase 2 Homo sapiens 87-91 29651325-1 2018 Irinotecan (CPT-11) is an anticancer prodrug that is activated by the carboxylesterase CES2 and has been approved for the treatment of many types of solid tumors, including colorectal cancer. Irinotecan 12-18 carboxylesterase 2 Homo sapiens 70-86 29651325-1 2018 Irinotecan (CPT-11) is an anticancer prodrug that is activated by the carboxylesterase CES2 and has been approved for the treatment of many types of solid tumors, including colorectal cancer. Irinotecan 12-18 carboxylesterase 2 Homo sapiens 87-91 29316651-0 2018 CPT-11-Induced Delayed Diarrhea Develops via Reduced Aquaporin-3 Expression in the Colon. Irinotecan 0-6 aquaporin 3 (Gill blood group) Rattus norvegicus 53-64 29316651-2 2018 In this study, we analyzed the pathogenic mechanism of CPT-11-induced delayed diarrhea by focusing on water channel aquaporin-3 (AQP3) in the colon. Irinotecan 55-61 aquaporin 3 (Gill blood group) Rattus norvegicus 116-127 29316651-2 2018 In this study, we analyzed the pathogenic mechanism of CPT-11-induced delayed diarrhea by focusing on water channel aquaporin-3 (AQP3) in the colon. Irinotecan 55-61 aquaporin 3 (Gill blood group) Rattus norvegicus 129-133 29316651-3 2018 When rats received CPT-11, the expression level of AQP3 was reduced during severe diarrhea. Irinotecan 19-25 aquaporin 3 (Gill blood group) Rattus norvegicus 51-55 29316651-5 2018 When celecoxib, an anti-inflammatory drug, was concomitantly administered, both the diarrhea and the reduced expression of AQP3 induced by CPT-11 were suppressed. Irinotecan 139-145 aquaporin 3 (Gill blood group) Rattus norvegicus 123-127 29316651-7 2018 These results showed that when CPT-11 is administered, the expression level of AQP3 in the colon is reduced, resulting in delayed diarrhea by preventing water transport from the intestinal tract. Irinotecan 31-37 aquaporin 3 (Gill blood group) Rattus norvegicus 79-83 29316651-8 2018 It was also suggested that the reduced expression of AQP3 might be due to the inflammation that occurs following the loss of colonic crypt cells and to the damage caused by the direct activation of macrophages by CPT-11. Irinotecan 213-219 aquaporin 3 (Gill blood group) Rattus norvegicus 53-57 29316651-9 2018 Therefore, it was considered that anti-inflammatory drugs that suppress the reduction of AQP3 expression could prevent CPT-11-induced delayed diarrhea. Irinotecan 119-125 aquaporin 3 (Gill blood group) Rattus norvegicus 89-93 30504681-7 2018 Vitamin C and/or irinotecan administration decreased the plasma level of ROS and IL-6 and increased the expression of collagen type I and caspase-1. Irinotecan 17-27 interleukin 6 Mus musculus 81-85 30504681-7 2018 Vitamin C and/or irinotecan administration decreased the plasma level of ROS and IL-6 and increased the expression of collagen type I and caspase-1. Irinotecan 17-27 caspase 1 Mus musculus 138-147 29210644-4 2018 CES2 plays crucial roles in the metabolic activation of many prodrugs including anticancer agents capecitabine and CPT-11. Irinotecan 115-121 carboxylesterase 2 Homo sapiens 0-4 29210644-5 2018 Co-administration with CES2 inhibitors may ameliorate CPT-11 associated lifethreatening diarrhea or improve the half-lives of CES2-substrate drugs. Irinotecan 54-60 carboxylesterase 2 Homo sapiens 23-27 31352923-5 2018 Compared with the free SN-38-BOC group, enhanced cytotoxicity and apoptotic induction were obtained from the SN-38-BOC micelle-treated group in both HCT116 and CT26 cells. Irinotecan 23-28 BOC cell adhesion associated, oncogene regulated Homo sapiens 115-118 31352923-7 2018 Furthermore, SN-38-BOC micelles exhibited stronger inhibition of tumor growth in both HCT116 and CT26 subcutaneous xenograft tumor models. Irinotecan 13-18 BOC cell adhesion associated, oncogene regulated Homo sapiens 19-22 31352923-8 2018 Histological analysis revealed that SN-38-BOC micelles showed a more effective anti-tumor activity than the free drug by inducing more apoptosis, inhibiting angiogenesis, and suppressing proliferation. Irinotecan 36-41 BOC cell adhesion associated, oncogene regulated Homo sapiens 42-45 31352923-9 2018 Thus, the pH-activatable SN-38-BOC micelle could serve as a promising candidate in colorectal tumor therapy. Irinotecan 25-30 BOC cell adhesion associated, oncogene regulated Homo sapiens 31-34 29125991-4 2018 The in vitro study evaluated the efficacy of high-dose SN-38 and quercetin combined with low-dose SN-38 on cell viability, apoptosis, and beta-catenin expression. Irinotecan 55-60 catenin beta 1 Homo sapiens 138-150 29125991-6 2018 AGS cells treated with a high dose of SN-38 exhibited up-regulation of beta-catenin protein expression, whereas quercetin-treated cells (either quercetin alone or combined with low-dose SN-38) exhibited lower protein levels of beta-catenin. Irinotecan 38-43 catenin beta 1 Homo sapiens 71-83 29125991-6 2018 AGS cells treated with a high dose of SN-38 exhibited up-regulation of beta-catenin protein expression, whereas quercetin-treated cells (either quercetin alone or combined with low-dose SN-38) exhibited lower protein levels of beta-catenin. Irinotecan 38-43 catenin beta 1 Homo sapiens 227-239 29125991-6 2018 AGS cells treated with a high dose of SN-38 exhibited up-regulation of beta-catenin protein expression, whereas quercetin-treated cells (either quercetin alone or combined with low-dose SN-38) exhibited lower protein levels of beta-catenin. Irinotecan 186-191 catenin beta 1 Homo sapiens 227-239 29125991-7 2018 In the AGS xenograft mouse model, gene expression of cyclooxygenase-2 and epithelial-mesenchymal transition-related markers, such as Twist1 and ITGbeta6, were lower in combined treatments with quercetin and low-dose irinotecan than high-dose irinotecan alone. Irinotecan 216-226 prostaglandin-endoperoxide synthase 2 Mus musculus 53-69 29125991-7 2018 In the AGS xenograft mouse model, gene expression of cyclooxygenase-2 and epithelial-mesenchymal transition-related markers, such as Twist1 and ITGbeta6, were lower in combined treatments with quercetin and low-dose irinotecan than high-dose irinotecan alone. Irinotecan 242-252 prostaglandin-endoperoxide synthase 2 Mus musculus 53-69 29125991-8 2018 Furthermore, the concentration of angiogenesis-associated factors (vascular endothelial growth factor (VEGF)-A and VEGF-receptor 2) and percentage of Tie2-expressing monocytes was significantly down-regulated in combined treatments with quercetin and irinotecan. Irinotecan 251-261 vascular endothelial growth factor A Homo sapiens 67-101 29125991-8 2018 Furthermore, the concentration of angiogenesis-associated factors (vascular endothelial growth factor (VEGF)-A and VEGF-receptor 2) and percentage of Tie2-expressing monocytes was significantly down-regulated in combined treatments with quercetin and irinotecan. Irinotecan 251-261 vascular endothelial growth factor A Homo sapiens 103-107 29125991-8 2018 Furthermore, the concentration of angiogenesis-associated factors (vascular endothelial growth factor (VEGF)-A and VEGF-receptor 2) and percentage of Tie2-expressing monocytes was significantly down-regulated in combined treatments with quercetin and irinotecan. Irinotecan 251-261 vascular endothelial growth factor A Homo sapiens 115-119 29125991-8 2018 Furthermore, the concentration of angiogenesis-associated factors (vascular endothelial growth factor (VEGF)-A and VEGF-receptor 2) and percentage of Tie2-expressing monocytes was significantly down-regulated in combined treatments with quercetin and irinotecan. Irinotecan 251-261 TEK receptor tyrosine kinase Homo sapiens 150-154 28834127-4 2018 Replacement of vincristine, irinotecan, and temozolomide (VIT) for VAC induced a marked tumor reduction and normalization of the miR-206 levels. Irinotecan 28-38 microRNA 206 Homo sapiens 129-136 29589315-7 2018 It has been established that UGT1A1*28 polymorphism is associated with irinotecan toxicity, but this test is rarely performed as the management strategy has not been identified. Irinotecan 71-81 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 29-35 29079710-0 2018 Selective and Concentrated Accretion of SN-38 with a CEACAM5-Targeting Antibody-Drug Conjugate (ADC), Labetuzumab Govitecan (IMMU-130). Irinotecan 40-45 CEA cell adhesion molecule 5 Homo sapiens 53-60 29138869-3 2018 Mutated p53 in CRC was reported to be associated with resistance to commonly used chemotherapeutic agents including, 5-fluorouracil, oxaliplatin and irinotecan. Irinotecan 149-159 tumor protein p53 Homo sapiens 8-11 29192320-0 2018 Combined SN-38 and gefitinib treatment promotes CD44 degradation in head and neck squamous cell carcinoma cells. Irinotecan 9-14 CD44 molecule (Indian blood group) Homo sapiens 48-52 29192320-2 2018 Among the tested drugs, we focused on combined SN-38, which is the active metabolite produced from irinotecan hydrochloride - a type I DNA topoisomerase inhibitor - after it is metabolized by carboxylesterase in the liver and gefitinib, an EGFR tyrosine kinase inhibitor treatment, based on the ability of this combination to inhibit HNSCC cell growth. Irinotecan 47-52 DNA topoisomerase I Homo sapiens 128-152 29192320-2 2018 Among the tested drugs, we focused on combined SN-38, which is the active metabolite produced from irinotecan hydrochloride - a type I DNA topoisomerase inhibitor - after it is metabolized by carboxylesterase in the liver and gefitinib, an EGFR tyrosine kinase inhibitor treatment, based on the ability of this combination to inhibit HNSCC cell growth. Irinotecan 99-123 DNA topoisomerase I Homo sapiens 128-152 29192320-6 2018 Next, we investigated the mechanism whereby SN-38 and gefitinib inhibited CD44 expression in vitro. Irinotecan 44-49 CD44 molecule (Indian blood group) Homo sapiens 74-78 29192320-8 2018 The present study revealed that combined gefitinib and SN-38 treatment inhibits CD44 expression by promoting its lysosomal degradation in HNSCC cells. Irinotecan 55-60 CD44 molecule (Indian blood group) Homo sapiens 80-84 29891024-10 2018 Previous reports, studies of recombinant UGT isoforms indicated that SN-38 glucuronidation was mainly catalyzed by UGT1A1. Irinotecan 69-74 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 41-44 29891024-10 2018 Previous reports, studies of recombinant UGT isoforms indicated that SN-38 glucuronidation was mainly catalyzed by UGT1A1. Irinotecan 69-74 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 115-121 30673201-4 2018 On the other hand, carriers of various genotypes of UGT1A1 differ significantly in metabolism characteristics of a number of common medications (irinotecan, belinostat, etc. Irinotecan 145-155 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 52-58 27845419-0 2018 ABC transporter polymorphisms are associated with irinotecan pharmacokinetics and neutropenia. Irinotecan 50-60 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 0-15 27845419-4 2018 rs6498588 (ABCC1) and rs12720066 (ABCB1) were associated with increased SN-38 exposure, and rs17501331 (ABCC1) and rs12720066 were associated with lower absolute neutrophil count nadir. Irinotecan 72-77 ATP binding cassette subfamily C member 1 Homo sapiens 11-16 27845419-4 2018 rs6498588 (ABCC1) and rs12720066 (ABCB1) were associated with increased SN-38 exposure, and rs17501331 (ABCC1) and rs12720066 were associated with lower absolute neutrophil count nadir. Irinotecan 72-77 ATP binding cassette subfamily B member 1 Homo sapiens 34-39 27845419-7 2018 These results suggest that genetic variation in ABCC1 and ABCB1 may contribute to irinotecan-induced neutropenia by altering expression of transporters involved in irinotecan metabolite disposition. Irinotecan 82-92 ATP binding cassette subfamily C member 1 Homo sapiens 48-53 27845419-7 2018 These results suggest that genetic variation in ABCC1 and ABCB1 may contribute to irinotecan-induced neutropenia by altering expression of transporters involved in irinotecan metabolite disposition. Irinotecan 82-92 ATP binding cassette subfamily B member 1 Homo sapiens 58-63 27845419-7 2018 These results suggest that genetic variation in ABCC1 and ABCB1 may contribute to irinotecan-induced neutropenia by altering expression of transporters involved in irinotecan metabolite disposition. Irinotecan 164-174 ATP binding cassette subfamily C member 1 Homo sapiens 48-53 29199543-4 2018 NR1I2 and NR1I3 genetic polymorphisms can affect the pharmacokinetics and therapeutic response to many drugs, such as irinotecan, tacrolimus and atazanavir. Irinotecan 118-128 nuclear receptor subfamily 1 group I member 2 Homo sapiens 0-5 27845419-7 2018 These results suggest that genetic variation in ABCC1 and ABCB1 may contribute to irinotecan-induced neutropenia by altering expression of transporters involved in irinotecan metabolite disposition. Irinotecan 164-174 ATP binding cassette subfamily B member 1 Homo sapiens 58-63 29019266-2 2018 The purpose of this study was to develop a thermo-sensitive hydrogel system with acidic SN-38 liposomes (SN-38-Lip-Gel) for local chemotherapy to solve these problems and to evaluate its antitumor activity and tissue distribution in tumor-bearing mice. Irinotecan 88-93 CCAAT/enhancer binding protein (C/EBP), beta Mus musculus 111-114 29019266-7 2018 SN-38-Lip-Gel suggested pH-dependent stability, the percentage of SN-38 A remaining decreased along with the increasing pH. Irinotecan 0-5 CCAAT/enhancer binding protein (C/EBP), beta Mus musculus 6-9 29019266-9 2018 In conclusion, SN-38-Lip-Gel could improve the effective use of SN-38 by stabilizing the lactone form, extending the drug release, providing a high local drug concentration, and reducing systemic toxicity. Irinotecan 15-20 CCAAT/enhancer binding protein (C/EBP), beta Mus musculus 21-24 29019266-9 2018 In conclusion, SN-38-Lip-Gel could improve the effective use of SN-38 by stabilizing the lactone form, extending the drug release, providing a high local drug concentration, and reducing systemic toxicity. Irinotecan 64-69 CCAAT/enhancer binding protein (C/EBP), beta Mus musculus 21-24 29199543-4 2018 NR1I2 and NR1I3 genetic polymorphisms can affect the pharmacokinetics and therapeutic response to many drugs, such as irinotecan, tacrolimus and atazanavir. Irinotecan 118-128 nuclear receptor subfamily 1 group I member 3 Homo sapiens 10-15 29052349-0 2018 Relationship between UGT1A1*27 and UGT1A1*7 polymorphisms and irinotecan-related toxicities in patients with lung cancer. Irinotecan 62-72 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 21-27 29052349-0 2018 Relationship between UGT1A1*27 and UGT1A1*7 polymorphisms and irinotecan-related toxicities in patients with lung cancer. Irinotecan 62-72 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 35-41 29052349-1 2018 BACKGROUND: The objective of this study was to evaluate the effects of gene polymorphisms, including UGT1A1*7, *27, and *29, on the safety of irinotecan therapy. Irinotecan 142-152 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 101-107 29052349-8 2018 SN-38 tended to remain in the blood for a prolonged period after the infusion of irinotecan in patients with UGT1A1*27 or UGT1A1*28 polymorphisms. Irinotecan 0-5 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 109-115 29052349-8 2018 SN-38 tended to remain in the blood for a prolonged period after the infusion of irinotecan in patients with UGT1A1*27 or UGT1A1*28 polymorphisms. Irinotecan 0-5 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 122-128 29052349-8 2018 SN-38 tended to remain in the blood for a prolonged period after the infusion of irinotecan in patients with UGT1A1*27 or UGT1A1*28 polymorphisms. Irinotecan 81-91 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 109-115 29052349-10 2018 CONCLUSION: UGT1A1*27 can occur separately from UGT1A1*28 and is related to leukopenia during irinotecan treatment. Irinotecan 94-104 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-18 29052349-10 2018 CONCLUSION: UGT1A1*27 can occur separately from UGT1A1*28 and is related to leukopenia during irinotecan treatment. Irinotecan 94-104 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 48-54 29052349-11 2018 UGT1A1*7 is less relevant to irinotecan-induced toxicities, and UGT1A1*29 seems to have little clinical impact. Irinotecan 29-39 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 29487697-1 2018 Our study addresses the issue of the clinical reliability of three candidate DPYD and one UGT single nucleotide polymorphisms in predicting 5-fluorouracil- and irinotecan-related adverse events. Irinotecan 160-170 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 90-93 29487697-5 2018 Present results support the preemptive screening of mentioned DPYD and UGT1A1 variants to identify patients at risk of clinically relevant 5-fluoruracil- and irinotecan-related AEs, in order to improve treatments" safety through a "genotype-guided" approach. Irinotecan 158-168 dihydropyrimidine dehydrogenase Homo sapiens 62-66 29487697-5 2018 Present results support the preemptive screening of mentioned DPYD and UGT1A1 variants to identify patients at risk of clinically relevant 5-fluoruracil- and irinotecan-related AEs, in order to improve treatments" safety through a "genotype-guided" approach. Irinotecan 158-168 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 71-77 29031818-0 2017 Camptothecin and its analog SN-38, the active metabolite of irinotecan, inhibit binding of the transcriptional regulator and oncoprotein FUBP1 to its DNA target sequence FUSE. Irinotecan 28-33 far upstream element binding protein 1 Homo sapiens 137-142 29277179-1 2017 BACKGROUND: Uridine-diphosphoglucuronosyl transferase 1A1 (UGT1A1), UGT1A1*28 polymorphism can reduce UGT1A1 enzymatic activity, which may lead to severe toxicities in patients who receive irinotecan. Irinotecan 189-199 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-57 29277179-1 2017 BACKGROUND: Uridine-diphosphoglucuronosyl transferase 1A1 (UGT1A1), UGT1A1*28 polymorphism can reduce UGT1A1 enzymatic activity, which may lead to severe toxicities in patients who receive irinotecan. Irinotecan 189-199 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 59-65 29277179-1 2017 BACKGROUND: Uridine-diphosphoglucuronosyl transferase 1A1 (UGT1A1), UGT1A1*28 polymorphism can reduce UGT1A1 enzymatic activity, which may lead to severe toxicities in patients who receive irinotecan. Irinotecan 189-199 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 68-74 29277179-1 2017 BACKGROUND: Uridine-diphosphoglucuronosyl transferase 1A1 (UGT1A1), UGT1A1*28 polymorphism can reduce UGT1A1 enzymatic activity, which may lead to severe toxicities in patients who receive irinotecan. Irinotecan 189-199 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 68-74 29031818-0 2017 Camptothecin and its analog SN-38, the active metabolite of irinotecan, inhibit binding of the transcriptional regulator and oncoprotein FUBP1 to its DNA target sequence FUSE. Irinotecan 28-33 Polykaryocytosis inducer Homo sapiens 170-174 29031818-0 2017 Camptothecin and its analog SN-38, the active metabolite of irinotecan, inhibit binding of the transcriptional regulator and oncoprotein FUBP1 to its DNA target sequence FUSE. Irinotecan 60-70 far upstream element binding protein 1 Homo sapiens 137-142 29031818-0 2017 Camptothecin and its analog SN-38, the active metabolite of irinotecan, inhibit binding of the transcriptional regulator and oncoprotein FUBP1 to its DNA target sequence FUSE. Irinotecan 60-70 Polykaryocytosis inducer Homo sapiens 170-174 29031818-3 2017 In this study, we screened an FDA-approved drug library and discovered that the Topoisomerase I (TOP1) inhibitor camptothecin (CPT) and its derivative 7-ethyl-10-hydroxycamptothecin (SN-38), the active irinotecan metabolite that is used in the clinics in combination with other chemotherapeutics to treat carcinoma, inhibit FUBP1 activity. Irinotecan 151-181 far upstream element binding protein 1 Homo sapiens 324-329 29031818-3 2017 In this study, we screened an FDA-approved drug library and discovered that the Topoisomerase I (TOP1) inhibitor camptothecin (CPT) and its derivative 7-ethyl-10-hydroxycamptothecin (SN-38), the active irinotecan metabolite that is used in the clinics in combination with other chemotherapeutics to treat carcinoma, inhibit FUBP1 activity. Irinotecan 183-188 far upstream element binding protein 1 Homo sapiens 324-329 29031818-5 2017 Our results suggest the interference with the FUBP1/FUSE interaction as a further molecular mechanism that, in addition to the inactivation of TOP1, may contribute to the therapeutic potential of CPT/SN-38. Irinotecan 200-205 far upstream element binding protein 1 Homo sapiens 46-51 29031818-5 2017 Our results suggest the interference with the FUBP1/FUSE interaction as a further molecular mechanism that, in addition to the inactivation of TOP1, may contribute to the therapeutic potential of CPT/SN-38. Irinotecan 200-205 Polykaryocytosis inducer Homo sapiens 52-56 29031818-6 2017 Targeting of FUBP1 in HCC therapy with SN-38/irinotecan could be a particularly interesting option because of the high FUBP1 levels in HCC cells and their dependency on FUBP1 expression. Irinotecan 39-44 far upstream element binding protein 1 Homo sapiens 13-18 29031818-6 2017 Targeting of FUBP1 in HCC therapy with SN-38/irinotecan could be a particularly interesting option because of the high FUBP1 levels in HCC cells and their dependency on FUBP1 expression. Irinotecan 39-44 far upstream element binding protein 1 Homo sapiens 119-124 29031818-6 2017 Targeting of FUBP1 in HCC therapy with SN-38/irinotecan could be a particularly interesting option because of the high FUBP1 levels in HCC cells and their dependency on FUBP1 expression. Irinotecan 39-44 far upstream element binding protein 1 Homo sapiens 119-124 29031818-6 2017 Targeting of FUBP1 in HCC therapy with SN-38/irinotecan could be a particularly interesting option because of the high FUBP1 levels in HCC cells and their dependency on FUBP1 expression. Irinotecan 45-55 far upstream element binding protein 1 Homo sapiens 13-18 29031818-6 2017 Targeting of FUBP1 in HCC therapy with SN-38/irinotecan could be a particularly interesting option because of the high FUBP1 levels in HCC cells and their dependency on FUBP1 expression. Irinotecan 45-55 far upstream element binding protein 1 Homo sapiens 119-124 29031818-6 2017 Targeting of FUBP1 in HCC therapy with SN-38/irinotecan could be a particularly interesting option because of the high FUBP1 levels in HCC cells and their dependency on FUBP1 expression. Irinotecan 45-55 far upstream element binding protein 1 Homo sapiens 119-124 29340025-0 2017 The novel ATM inhibitor (AZ31) enhances antitumor activity in patient derived xenografts that are resistant to irinotecan monotherapy. Irinotecan 111-121 ATM serine/threonine kinase Homo sapiens 10-13 28940490-6 2017 KEY FINDINGS: Eight amino acids residues in TRPA1 established stable interactions with carvacryl acetate, which led to pharmacological efficacy against CPT-11-induced intestinal mucositis via reduction of both neutropenia and bacteremia, increase in villi height and crypt depth, decrease in pro-inflammatory cytokines (interleukin-1beta, keratinocyte chemoattractant and tumour necrosis factor-alpha) and decrease in malondialdehyde and nitric oxide metabolite levels in the jejunum. Irinotecan 152-158 transient receptor potential cation channel, subfamily A, member 1 Mus musculus 44-49 29312810-8 2017 Thus, the sequential treatment with SN-38 followed by FTD may be useful for the combination therapy of FTD/TPI and CPT-11 against relapsed colorectal cancer after 5-FU-based chemotherapy. Irinotecan 36-41 triosephosphate isomerase 1 Homo sapiens 107-110 29340025-3 2017 The objective of this preclinical study was to determine whether ATM inhibition would enhance sensitivity to irinotecan treatment. Irinotecan 109-119 ATM serine/threonine kinase Homo sapiens 65-68 29340025-8 2017 Treatment with irinotecan significantly elevated the ATM pathway evident by an increase in the activation of H2AX and RAD50. Irinotecan 15-25 ATM serine/threonine kinase Homo sapiens 53-56 29340025-8 2017 Treatment with irinotecan significantly elevated the ATM pathway evident by an increase in the activation of H2AX and RAD50. Irinotecan 15-25 H2A.X variant histone Homo sapiens 109-113 29340025-8 2017 Treatment with irinotecan significantly elevated the ATM pathway evident by an increase in the activation of H2AX and RAD50. Irinotecan 15-25 RAD50 double strand break repair protein Homo sapiens 118-123 29340025-9 2017 Combinational therapy reduced the activation of H2AX and RAD50 when compared to irinotecan alone in the combination sensitive CRC098. Irinotecan 80-90 H2A.X variant histone Homo sapiens 48-52 29174536-3 2017 Irinotecan and etoposide inhibited the uptake of [3H]-uridine and [3H]-DAC at 10 s and 5 min, while cytarabine and gemcitabine only inhibited that of [3H]-DAC at 5 min. Irinotecan 0-10 arylacetamide deacetylase Homo sapiens 71-74 29174536-3 2017 Irinotecan and etoposide inhibited the uptake of [3H]-uridine and [3H]-DAC at 10 s and 5 min, while cytarabine and gemcitabine only inhibited that of [3H]-DAC at 5 min. Irinotecan 0-10 arylacetamide deacetylase Homo sapiens 155-158 29174536-4 2017 Irinotecan and etoposide inhibited [3H]-DAC uptake in negative control small interfering RNA (siRNA)- or dCK siRNA-transfected cells at 10 s, whereas cytarabine and gemcitabine did not. Irinotecan 0-10 arylacetamide deacetylase Homo sapiens 40-43 29174536-4 2017 Irinotecan and etoposide inhibited [3H]-DAC uptake in negative control small interfering RNA (siRNA)- or dCK siRNA-transfected cells at 10 s, whereas cytarabine and gemcitabine did not. Irinotecan 0-10 Calcium/calmodulin-dependent protein kinase II Drosophila melanogaster 105-108 28940490-6 2017 KEY FINDINGS: Eight amino acids residues in TRPA1 established stable interactions with carvacryl acetate, which led to pharmacological efficacy against CPT-11-induced intestinal mucositis via reduction of both neutropenia and bacteremia, increase in villi height and crypt depth, decrease in pro-inflammatory cytokines (interleukin-1beta, keratinocyte chemoattractant and tumour necrosis factor-alpha) and decrease in malondialdehyde and nitric oxide metabolite levels in the jejunum. Irinotecan 152-158 interleukin 1 beta Mus musculus 320-337 28940490-7 2017 CONCLUSIONS: Carvacryl acetate is a promising anti-inflammatory and antioxidant agent, a fact confirmed through observations of its interactions with TRPA1 in CPT-11-induced intestinal mucositis in mice. Irinotecan 159-165 transient receptor potential cation channel, subfamily A, member 1 Mus musculus 150-155 28948511-4 2017 The involvement of ABCG2 transporter in irinotecan resistance has been established. Irinotecan 40-50 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 19-24 28801072-9 2017 Oxidative stress parameters and intracellular mediators as well as the MPO activity were determined in intestinal mucosa by colorimetric methods Our result indicated that irinotecan produces an intestinal fluid accumulation and electrolyte transport disorders. Irinotecan 171-181 myeloperoxidase Rattus norvegicus 71-74 29344244-6 2017 Furthermore, PSAT1 overexpression was associated with response to irinotecan, 5-fluorouracil and leucovorin chemotherapy, and with shorter survival time, and retained significance as an independent prognostic factor for CRC when subjected to the multivariate analysis with a Cox"s proportional hazards model. Irinotecan 66-76 phosphoserine aminotransferase 1 Homo sapiens 13-18 28948511-5 2017 The current study was designed to characterize SN38-loaded pegylated (polyethylene glycol) PLGA [poly(lactic-co-glycolic acid)]-verapamil nanoparticles (NPs), and to distinguish the cytotoxic effect of SN38-PEG-PLGA-Ver NPs and the ability of SN38-PEG-PLGA-Ver NPs to inhibit drug resistance through the inhibition of ABCG2 expression. Irinotecan 47-51 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 318-323 29184868-1 2017 We describe an extremely rare case of advanced pure primary ovarian squamous cell carcinoma (SCC), treated by adjuvant chemotherapy with dose-dense paclitaxel combined with carboplatin (dd-TC) plus the combination chemotherapy with irinotecan and cisplatin (CPT-P), with long-term recurrence-free survival. Irinotecan 232-242 serpin family B member 3 Homo sapiens 93-96 29067643-5 2017 The MTD for the combination of gedatolisib with irinotecan 180 mg/m2 was estimated to be 110 mg weekly and for the combination with PD-0325901 was not reached at the highest dose evaluated (gedatolisib 154 mg weekly). Irinotecan 48-58 metallothionein 1E Homo sapiens 4-7 29180337-3 2017 Western blotting was performed to analyze the effect of bFGF-mAb combined with irinotecan on AKT and ERK1/2 phosphorylation in the cells. Irinotecan 79-89 AKT serine/threonine kinase 1 Homo sapiens 93-96 29180337-3 2017 Western blotting was performed to analyze the effect of bFGF-mAb combined with irinotecan on AKT and ERK1/2 phosphorylation in the cells. Irinotecan 79-89 mitogen-activated protein kinase 3 Homo sapiens 101-107 29180337-5 2017 The inhibitory rate was significantly higher in H223 cells treated with bFGF mAb + irinotecan (54.30%) than in cell treated with bFGF mAb (18.73%) or irinotecan (21.96%) alone (P<0.05). Irinotecan 83-93 fibroblast growth factor 2 Homo sapiens 72-76 29180337-6 2017 Both bFGF mAb and irinotecan induced H223 cell apoptosis in a dose-dependent manner (P<0.05), and the combined treatment resulted in a significantly higher early apoptosis rates (6.5%) than treatment with bFGF mAb (2.7%) or irinotecan (4.3%) alone (P<0.05). Irinotecan 18-28 fibroblast growth factor 2 Homo sapiens 208-212 29180337-6 2017 Both bFGF mAb and irinotecan induced H223 cell apoptosis in a dose-dependent manner (P<0.05), and the combined treatment resulted in a significantly higher early apoptosis rates (6.5%) than treatment with bFGF mAb (2.7%) or irinotecan (4.3%) alone (P<0.05). Irinotecan 227-237 fibroblast growth factor 2 Homo sapiens 5-9 29180337-7 2017 bFGF mAb and irinotecan, either alone or in combination, significantly inhibited the levels of p-AKT protein and p-ERK1/2 protein without obviously affecting AKT and ERK1/2 protein levels. Irinotecan 13-23 AKT serine/threonine kinase 1 Homo sapiens 97-100 29180337-7 2017 bFGF mAb and irinotecan, either alone or in combination, significantly inhibited the levels of p-AKT protein and p-ERK1/2 protein without obviously affecting AKT and ERK1/2 protein levels. Irinotecan 13-23 mitogen-activated protein kinase 3 Homo sapiens 115-121 29180337-7 2017 bFGF mAb and irinotecan, either alone or in combination, significantly inhibited the levels of p-AKT protein and p-ERK1/2 protein without obviously affecting AKT and ERK1/2 protein levels. Irinotecan 13-23 AKT serine/threonine kinase 1 Homo sapiens 158-161 29180337-7 2017 bFGF mAb and irinotecan, either alone or in combination, significantly inhibited the levels of p-AKT protein and p-ERK1/2 protein without obviously affecting AKT and ERK1/2 protein levels. Irinotecan 13-23 mitogen-activated protein kinase 3 Homo sapiens 166-172 29180337-8 2017 CONCLUSION: bFGF mAb and irinotecan produce synergistic inhibitory effects on small cell lung cancer H223 cells by suppressing proliferation and promoting apoptosis of the cells, and can effectively block the MAPK/ERK and PI3K/AKT signaling pathways associated with bFGF. Irinotecan 25-35 mitogen-activated protein kinase 3 Homo sapiens 209-213 29180337-8 2017 CONCLUSION: bFGF mAb and irinotecan produce synergistic inhibitory effects on small cell lung cancer H223 cells by suppressing proliferation and promoting apoptosis of the cells, and can effectively block the MAPK/ERK and PI3K/AKT signaling pathways associated with bFGF. Irinotecan 25-35 mitogen-activated protein kinase 1 Homo sapiens 214-217 29180337-8 2017 CONCLUSION: bFGF mAb and irinotecan produce synergistic inhibitory effects on small cell lung cancer H223 cells by suppressing proliferation and promoting apoptosis of the cells, and can effectively block the MAPK/ERK and PI3K/AKT signaling pathways associated with bFGF. Irinotecan 25-35 AKT serine/threonine kinase 1 Homo sapiens 227-230 29180337-8 2017 CONCLUSION: bFGF mAb and irinotecan produce synergistic inhibitory effects on small cell lung cancer H223 cells by suppressing proliferation and promoting apoptosis of the cells, and can effectively block the MAPK/ERK and PI3K/AKT signaling pathways associated with bFGF. Irinotecan 25-35 fibroblast growth factor 2 Homo sapiens 266-270 29016119-5 2017 The best compound was active in a very low micromolar concentration range against all three transporters and restored sensitivity toward daunorubicin (P-gp and MRP1) and SN-38 (BCRP) in A2780/ADR (P-gp), H69AR (MRP1), and MDCK II BCRP (BCRP) cells. Irinotecan 170-175 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 177-181 29016119-5 2017 The best compound was active in a very low micromolar concentration range against all three transporters and restored sensitivity toward daunorubicin (P-gp and MRP1) and SN-38 (BCRP) in A2780/ADR (P-gp), H69AR (MRP1), and MDCK II BCRP (BCRP) cells. Irinotecan 170-175 ATP binding cassette subfamily B member 1 Homo sapiens 197-201 29016119-5 2017 The best compound was active in a very low micromolar concentration range against all three transporters and restored sensitivity toward daunorubicin (P-gp and MRP1) and SN-38 (BCRP) in A2780/ADR (P-gp), H69AR (MRP1), and MDCK II BCRP (BCRP) cells. Irinotecan 170-175 ATP binding cassette subfamily C member 1 Homo sapiens 211-215 29016119-5 2017 The best compound was active in a very low micromolar concentration range against all three transporters and restored sensitivity toward daunorubicin (P-gp and MRP1) and SN-38 (BCRP) in A2780/ADR (P-gp), H69AR (MRP1), and MDCK II BCRP (BCRP) cells. Irinotecan 170-175 ATP binding cassette subfamily G member 2 Canis lupus familiaris 222-234 29016119-5 2017 The best compound was active in a very low micromolar concentration range against all three transporters and restored sensitivity toward daunorubicin (P-gp and MRP1) and SN-38 (BCRP) in A2780/ADR (P-gp), H69AR (MRP1), and MDCK II BCRP (BCRP) cells. Irinotecan 170-175 ATP binding cassette subfamily G member 2 Canis lupus familiaris 230-234 28904007-7 2017 In addition, when used in cytotoxicity assays, GO-Y030 and GO-Y078 were found to improve the sensitivity of the anticancer drug, SN-38, in K562/BCRP cells. Irinotecan 129-134 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 144-148 29113203-1 2017 Uridine diphosphate glucuronosyltransferase 1A (UGT1A1), which affects irinotecan metabolism, has been associated with severe adverse reactions in patients with cancer treated with irinotecan. Irinotecan 71-81 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 48-54 29113203-1 2017 Uridine diphosphate glucuronosyltransferase 1A (UGT1A1), which affects irinotecan metabolism, has been associated with severe adverse reactions in patients with cancer treated with irinotecan. Irinotecan 181-191 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 48-54 29113203-9 2017 These findings suggested that the UGT1A1*28 and UGT1A1*6 genotypes may be associated with irinotecan-induced severe toxicity, and clarified the clinical importance of UGT1A1 polymorphisms, particularly UGT1A1*6, regarding irinotecan therapy in Chinese patients. Irinotecan 90-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 34-40 29113203-9 2017 These findings suggested that the UGT1A1*28 and UGT1A1*6 genotypes may be associated with irinotecan-induced severe toxicity, and clarified the clinical importance of UGT1A1 polymorphisms, particularly UGT1A1*6, regarding irinotecan therapy in Chinese patients. Irinotecan 90-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 48-54 29113203-9 2017 These findings suggested that the UGT1A1*28 and UGT1A1*6 genotypes may be associated with irinotecan-induced severe toxicity, and clarified the clinical importance of UGT1A1 polymorphisms, particularly UGT1A1*6, regarding irinotecan therapy in Chinese patients. Irinotecan 90-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 48-54 29113203-9 2017 These findings suggested that the UGT1A1*28 and UGT1A1*6 genotypes may be associated with irinotecan-induced severe toxicity, and clarified the clinical importance of UGT1A1 polymorphisms, particularly UGT1A1*6, regarding irinotecan therapy in Chinese patients. Irinotecan 90-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 48-54 29113203-9 2017 These findings suggested that the UGT1A1*28 and UGT1A1*6 genotypes may be associated with irinotecan-induced severe toxicity, and clarified the clinical importance of UGT1A1 polymorphisms, particularly UGT1A1*6, regarding irinotecan therapy in Chinese patients. Irinotecan 222-232 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 34-40 29163158-11 2017 And the underlying mechanism of drug interaction between CPT-11 and SXD involves decreasing hepatic Mrp-2 and P-gp expression and altering the activities of CES and UGT. Irinotecan 57-63 ATP binding cassette subfamily C member 2 Rattus norvegicus 100-105 29163158-11 2017 And the underlying mechanism of drug interaction between CPT-11 and SXD involves decreasing hepatic Mrp-2 and P-gp expression and altering the activities of CES and UGT. Irinotecan 57-63 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 110-114 29066969-0 2017 Improved Progression-Free Survival in Irinotecan-Treated Metastatic Colorectal Cancer Patients Carrying the HNF1A Coding Variant p.I27L. Irinotecan 38-48 HNF1 homeobox A Homo sapiens 108-113 29072417-1 2017 Irinotecan life threatening toxicity is partly related to cytotoxic drugmetabolite which is primarily inactivated by the UGT1A1 enzyme. Irinotecan 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 121-127 29072417-2 2017 The primary aim of the present research was tofind the correlation between UGT1A1-genotype and clinical toxicity of irinotecan. Irinotecan 116-126 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 75-81 29072417-13 2017 Conclusions: UGT1A1 28*/28* is strongly associatedwith drug"s life-threatening toxicity even in a moderate dose of Irinotecan. Irinotecan 115-125 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 13-19 28881286-6 2017 Moreover, via its effects on NF-kappaB, S06 effectively enhanced the sensitivity of the gastric cancer cells to irinotecan. Irinotecan 112-122 nuclear factor kappa B subunit 1 Homo sapiens 29-38 28817371-1 2017 Purpose The objectives were to evaluate dosing schedules of labetuzumab govitecan, an antibody-drug conjugate targeting carcinoembryonic antigen-related cell adhesion molecule 5 (CEACAM5) for tumor delivery of 7-ethyl-10-hydroxycamptothecin (SN-38), in an expanded phase II trial of patients with relapsed or refractory metastatic colorectal cancer. Irinotecan 210-240 CEA cell adhesion molecule 5 Homo sapiens 179-186 29066969-9 2017 Our findings suggest that the HNF1A rs2244608, or the tightly linked functional coding variant p.I27L, might be a potential prognostic marker with irinotecan-based regimens. Irinotecan 147-157 HNF1 homeobox A Homo sapiens 30-35 28679770-1 2017 Purpose: We evaluated a Trop-2-targeting antibody conjugated with SN-38 in metastatic small cell lung cancer (mSCLC) patients.Experimental Design: Sacituzumab govitecan was studied in patients with pretreated (median, 2; range, 1-7) mSCLC who received either 8 or 10 mg/kg i.v. Irinotecan 66-71 tumor associated calcium signal transducer 2 Homo sapiens 24-30 28878234-4 2017 Here, we identified a novel relationship between IL-7R signaling and steroid-resistance, and showed that an anti-IL-7R antibody conjugated with SN-38 (A7R-ADC-SN-38) has strong anti-tumor effects against both parental and steroid-resistant malignant cells. Irinotecan 144-149 interleukin 7 receptor Mus musculus 49-54 28421347-11 2017 CPT-11 caused a significant increase in the expression of TLR4, IL-6, TNF-alpha, IL-1beta and caspase-3 mRNAs in the large intestine. Irinotecan 0-6 toll-like receptor 4 Rattus norvegicus 58-62 28421347-11 2017 CPT-11 caused a significant increase in the expression of TLR4, IL-6, TNF-alpha, IL-1beta and caspase-3 mRNAs in the large intestine. Irinotecan 0-6 interleukin 6 Rattus norvegicus 64-68 28421347-11 2017 CPT-11 caused a significant increase in the expression of TLR4, IL-6, TNF-alpha, IL-1beta and caspase-3 mRNAs in the large intestine. Irinotecan 0-6 tumor necrosis factor Rattus norvegicus 70-79 28421347-11 2017 CPT-11 caused a significant increase in the expression of TLR4, IL-6, TNF-alpha, IL-1beta and caspase-3 mRNAs in the large intestine. Irinotecan 0-6 interleukin 1 beta Rattus norvegicus 81-89 28421347-11 2017 CPT-11 caused a significant increase in the expression of TLR4, IL-6, TNF-alpha, IL-1beta and caspase-3 mRNAs in the large intestine. Irinotecan 0-6 caspase 3 Rattus norvegicus 94-103 29262551-0 2017 The combination of temozolomide-irinotecan regresses a doxorubicin-resistant patient-derived orthotopic xenograft (PDOX) nude-mouse model of recurrent Ewing"s sarcoma with a FUS-ERG fusion and CDKN2A deletion: Direction for third-line patient therapy. Irinotecan 32-42 fused in sarcoma Mus musculus 174-177 29262551-0 2017 The combination of temozolomide-irinotecan regresses a doxorubicin-resistant patient-derived orthotopic xenograft (PDOX) nude-mouse model of recurrent Ewing"s sarcoma with a FUS-ERG fusion and CDKN2A deletion: Direction for third-line patient therapy. Irinotecan 32-42 cyclin dependent kinase inhibitor 2A Mus musculus 193-199 29066704-1 2017 We report 2 unusual cases of Stage IV rectal cancer for which pathological complete response was achieved with neoadjuvant chemotherapy(NAC)consisting of a combination of 5-fluorouracil(5-FU), oxaliplatin, Leucovorin(mFOLFOX6)or irinotecan(FOLFIRI), and bevacizumab, without radiotherapy. Irinotecan 229-239 X-linked Kx blood group Homo sapiens 136-139 28880238-0 2017 Implications of ABCG2 Expression on Irinotecan Treatment of Colorectal Cancer Patients: A Review. Irinotecan 36-46 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 16-21 28880238-3 2017 The ATP-binding cassette (ABC) transporter ABCG2/breast cancer resistance protein (BRCP) through its function in xenobiotic clearance might play an important role in irinotecan resistance. Irinotecan 166-176 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 43-48 28880238-3 2017 The ATP-binding cassette (ABC) transporter ABCG2/breast cancer resistance protein (BRCP) through its function in xenobiotic clearance might play an important role in irinotecan resistance. Irinotecan 166-176 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 49-81 28880238-4 2017 With a goal to evaluate the clinical significance of ABCG2 measurements, we here review the current literature on ABCG2 in relation to irinotecan treatment in CRC patients. Irinotecan 135-145 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 114-119 28880238-11 2017 In contrast, our recent exploratory study of ABCG2 mRNA expression in 580 patients with stage III primary CRC (subgroup from the randomized PETACC-3 study) indicated that high ABCG2 tumor tissue mRNA expression might be predictive for lack of efficacy of irinotecan. Irinotecan 255-265 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 176-181 28880238-12 2017 CONCLUSION: The biological role of ABCG2 in predicting clinical irinotecan sensitivity/resistance in CRC is uncertain. Irinotecan 64-74 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 35-40 28878234-4 2017 Here, we identified a novel relationship between IL-7R signaling and steroid-resistance, and showed that an anti-IL-7R antibody conjugated with SN-38 (A7R-ADC-SN-38) has strong anti-tumor effects against both parental and steroid-resistant malignant cells. Irinotecan 144-149 interleukin 7 receptor Mus musculus 113-118 28878234-4 2017 Here, we identified a novel relationship between IL-7R signaling and steroid-resistance, and showed that an anti-IL-7R antibody conjugated with SN-38 (A7R-ADC-SN-38) has strong anti-tumor effects against both parental and steroid-resistant malignant cells. Irinotecan 159-164 interleukin 7 receptor Mus musculus 49-54 28878234-4 2017 Here, we identified a novel relationship between IL-7R signaling and steroid-resistance, and showed that an anti-IL-7R antibody conjugated with SN-38 (A7R-ADC-SN-38) has strong anti-tumor effects against both parental and steroid-resistant malignant cells. Irinotecan 159-164 interleukin 7 receptor Mus musculus 113-118 28735070-0 2017 Suppression of the ATP-binding cassette transporter ABCC4 impairs neuroblastoma tumour growth and sensitises to irinotecan in vivo. Irinotecan 112-122 ATP binding cassette subfamily C member 4 Homo sapiens 52-57 28711984-3 2017 Recently, the efficacy of combining CPT-11 and anti-epidermal growth factor receptor (EGFR) agents was confirmed in patients with KRAS wild-type metastatic colorectal cancer. Irinotecan 36-42 KRAS proto-oncogene, GTPase Homo sapiens 130-134 28735070-5 2017 Loss of Abcc4 in the Th-MYCN transgenic neuroblastoma mouse model did not impact tumour formation; however, Abcc4-null neuroblastomas were strongly sensitised to the ABCC4 substrate drug irinotecan. Irinotecan 187-197 ATP-binding cassette, sub-family C (CFTR/MRP), member 4 Mus musculus 108-113 28735070-5 2017 Loss of Abcc4 in the Th-MYCN transgenic neuroblastoma mouse model did not impact tumour formation; however, Abcc4-null neuroblastomas were strongly sensitised to the ABCC4 substrate drug irinotecan. Irinotecan 187-197 ATP-binding cassette, sub-family C (CFTR/MRP), member 4 Mus musculus 166-171 28894577-3 2017 The aim of the present study was to assess the feasibility and efficacy of irinotecan and cisplatin in the management of patients with vaginal squamous cell cancer (SCC). Irinotecan 75-85 serpin family B member 3 Homo sapiens 165-168 28894577-11 2017 Thus, neoadjuvant chemotherapy with irinotecan combined with cisplatin is a feasible treatment for patients with early-stage vaginal SCC. Irinotecan 36-46 serpin family B member 3 Homo sapiens 133-136 28850174-24 2017 For HER-2 negative people, all different two-and three-drug combinations including irinotecan, docetaxel, oxaliplatin or oral 5-FU prodrugs are valid treatment options for advanced gastric cancer, and consideration of the side effects of each regimen is essential in the treatment decision. Irinotecan 83-93 erb-b2 receptor tyrosine kinase 2 Homo sapiens 4-9 28627611-0 2017 Epithelial-mesenchymal transition and cancer stem cell-like phenotype induced by Twist1 contribute to acquired resistance to irinotecan in colon cancer. Irinotecan 125-135 twist family bHLH transcription factor 1 Homo sapiens 81-87 28548889-0 2017 Therapy of Advanced Non-Small-Cell Lung Cancer With an SN-38-Anti-Trop-2 Drug Conjugate, Sacituzumab Govitecan. Irinotecan 55-60 tumor associated calcium signal transducer 2 Homo sapiens 66-72 28548889-2 2017 We studied sacituzumab govitecan (IMMU-132), a Trop-2 ADC, for the targeting of SN-38. Irinotecan 80-85 tumor associated calcium signal transducer 2 Homo sapiens 47-53 28627611-5 2017 In the present study, we further investigated how Twist1 contribute to acquired resistance to irinotecan in colon cancer. Irinotecan 94-104 twist family bHLH transcription factor 1 Homo sapiens 50-56 28627611-10 2017 Results showed that the LoVo/Twist1 cells perform a distinctly decreased sensitivity to irinotecan, downregulated expression of E-cadherin, upregulated expression of cluster of differentiation 44 (CD44), and a significant enhancement of invasion and migration potential by regulation of MMP2 compared with control cells. Irinotecan 88-98 twist family bHLH transcription factor 1 Homo sapiens 29-35 28627611-11 2017 In contrast, the inhibition of Twist1 transfected with siRNA could enhance the irinotecan sensitivity in LoVo/CPT-11R cells and downregulate the expression of vimentin and CD44. Irinotecan 79-89 twist family bHLH transcription factor 1 Homo sapiens 31-37 28627611-12 2017 Our data provide evidence that EMT and CSC-like phenotype induced by Twist1 contribute to acquire resistance to irinotecan and enhanced migration and invasion in colon cancer. Irinotecan 112-122 twist family bHLH transcription factor 1 Homo sapiens 69-75 28120346-6 2017 Cytotoxicity studies show that compound 7d sensitizes the ABCG2-overexpressing cells to chemotherapeutic drugs mitoxantrone and SN-38, which are well-established substrates of the ABCG2 transporter. Irinotecan 128-133 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 58-63 28120346-6 2017 Cytotoxicity studies show that compound 7d sensitizes the ABCG2-overexpressing cells to chemotherapeutic drugs mitoxantrone and SN-38, which are well-established substrates of the ABCG2 transporter. Irinotecan 128-133 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 180-185 28397089-5 2017 Of these, 19 sets gave relatively good description of the effect of UGT1A1 *28 and SLCO1B1 c.521T>C polymorphism on the SN-38 plasma concentration, neutropenia, and diarrhea observed in clinical studies reported mainly by Teft et al. Irinotecan 123-128 solute carrier organic anion transporter family member 1B1 Homo sapiens 83-90 28542923-0 2017 The polysialic acid mimetics idarubicin and irinotecan stimulate neuronal survival and neurite outgrowth and signal via protein kinase C. Polysialic acid (PSA) is a large, negatively charged, linear homopolymer of alpha2-8-linked sialic acid residues. Irinotecan 44-54 immunoglobulin binding protein 1 Homo sapiens 214-222 28542923-6 2017 Idarubicin and irinotecan stimulate neurite outgrowth and survival of cultured cerebellar neurons after oxidative stress via protein kinase C and Erk1/2 in a similar manner as colominic acid, whereas Fyn, casein kinase II and the phosphatase and tensin homolog are only involved in idarubicin and irinotecan-stimulated neurite outgrowth. Irinotecan 15-25 mitogen-activated protein kinase 3 Homo sapiens 146-152 28542923-6 2017 Idarubicin and irinotecan stimulate neurite outgrowth and survival of cultured cerebellar neurons after oxidative stress via protein kinase C and Erk1/2 in a similar manner as colominic acid, whereas Fyn, casein kinase II and the phosphatase and tensin homolog are only involved in idarubicin and irinotecan-stimulated neurite outgrowth. Irinotecan 15-25 FYN proto-oncogene, Src family tyrosine kinase Homo sapiens 200-203 28397089-9 2017 CONCLUSION: The VCS confirmed the importance of genetic polymorphisms of UGT1A1 *28 and SLCO1B1 c.521T>C in the irinotecan induced side effects. Irinotecan 115-125 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 73-79 28397089-9 2017 CONCLUSION: The VCS confirmed the importance of genetic polymorphisms of UGT1A1 *28 and SLCO1B1 c.521T>C in the irinotecan induced side effects. Irinotecan 115-125 solute carrier organic anion transporter family member 1B1 Homo sapiens 88-95 28749961-13 2017 In addition, FGF9 was also associated with irinotecan resistance and poor disease free survival. Irinotecan 43-53 fibroblast growth factor 9 Homo sapiens 13-17 28749961-15 2017 CONCLUSION: Targeting the MAPK signal transduction pathway through the targeting of the FGF2, FGF9, MECOM, PLA2G4C and PRKACB might increase tumor responsiveness to irinotecan treatment. Irinotecan 165-175 fibroblast growth factor 2 Homo sapiens 88-92 28749961-15 2017 CONCLUSION: Targeting the MAPK signal transduction pathway through the targeting of the FGF2, FGF9, MECOM, PLA2G4C and PRKACB might increase tumor responsiveness to irinotecan treatment. Irinotecan 165-175 fibroblast growth factor 9 Homo sapiens 94-98 28749961-15 2017 CONCLUSION: Targeting the MAPK signal transduction pathway through the targeting of the FGF2, FGF9, MECOM, PLA2G4C and PRKACB might increase tumor responsiveness to irinotecan treatment. Irinotecan 165-175 MDS1 and EVI1 complex locus Homo sapiens 100-105 28749961-15 2017 CONCLUSION: Targeting the MAPK signal transduction pathway through the targeting of the FGF2, FGF9, MECOM, PLA2G4C and PRKACB might increase tumor responsiveness to irinotecan treatment. Irinotecan 165-175 phospholipase A2 group IVC Homo sapiens 107-114 28749961-15 2017 CONCLUSION: Targeting the MAPK signal transduction pathway through the targeting of the FGF2, FGF9, MECOM, PLA2G4C and PRKACB might increase tumor responsiveness to irinotecan treatment. Irinotecan 165-175 protein kinase cAMP-activated catalytic subunit beta Homo sapiens 119-125 28526406-4 2017 However, irinotecan-resistant SCLC cells, such as SBC-3/SN-38, were refractory to trastuzumab despite high HER2 expression. Irinotecan 9-19 erb-b2 receptor tyrosine kinase 2 Homo sapiens 107-111 28207160-0 2017 Pharmacokinetic interactions in mice between irinotecan and MBL-II-141, an ABCG2 inhibitor. Irinotecan 45-55 ATP binding cassette subfamily G member 2 (Junior blood group) Mus musculus 75-80 28526406-6 2017 Treatment with T-DM1 significantly suppressed the growth of SBC-3/SN-38 xenografts in mice compared with trastuzumab (P < 0.05). Irinotecan 66-71 immunoglobulin heavy diversity 1-7 Homo sapiens 17-20 27737534-0 2017 ERCC1 Expression-Based Randomized Phase II Study of Gemcitabine/Cisplatin Versus Irinotecan/Cisplatin in Patients with Advanced Non-small Cell Lung Cancer. Irinotecan 81-91 ERCC excision repair 1, endonuclease non-catalytic subunit Homo sapiens 0-5 28585035-0 2017 UGT1A1*6 polymorphisms are correlated with irinotecan-induced neutropenia: a systematic review and meta-analysis. Irinotecan 43-53 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 28474802-0 2017 Rapid and sensitive detection of UGT1A1 polymorphisms associated with irinotecan toxicity by a novel DNA microarray. Irinotecan 70-80 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-39 28474802-4 2017 It is established that the polymorphisms of UGT1A1*28 and UGT1A1*6 are significantly associated with severe toxicity induced by the anti-cancer drug irinotecan. Irinotecan 149-159 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 44-50 28474802-4 2017 It is established that the polymorphisms of UGT1A1*28 and UGT1A1*6 are significantly associated with severe toxicity induced by the anti-cancer drug irinotecan. Irinotecan 149-159 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 58-64 28074472-0 2017 Cost Evaluation of Irinotecan-Related Toxicities Associated With the UGT1A1*28 Patient Genotype. Irinotecan 19-29 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 69-75 28677646-5 2017 EC50 values of 6-mercaptopurine or 7-Ethyl-10-hydroxy-camptothecin (SN-38) against cells expressing ABCC4 (WT) were also 1.4-2.0- or 1.9-fold higher than those against cells expressing the SNP variants of ABCC4 (K304N or E757K) or (K304N; P403L or E757K); respectively. Irinotecan 68-73 ATP binding cassette subfamily C member 4 Homo sapiens 100-105 28677646-5 2017 EC50 values of 6-mercaptopurine or 7-Ethyl-10-hydroxy-camptothecin (SN-38) against cells expressing ABCC4 (WT) were also 1.4-2.0- or 1.9-fold higher than those against cells expressing the SNP variants of ABCC4 (K304N or E757K) or (K304N; P403L or E757K); respectively. Irinotecan 68-73 ATP binding cassette subfamily C member 4 Homo sapiens 205-210 28074472-1 2017 The adoption of a preemptive UGT1A1*28 genotyping to increase irinotecan safety in clinical practice is still limited. Irinotecan 62-72 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 29-35 28637434-1 2017 BACKGROUND: To evaluate a new UGT1A and DPYD polymorphism panel to better predict irinotecan-induced toxicity and the clinical response in Chinese patients with metastatic colorectal cancer (mCRC). Irinotecan 82-92 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 30-35 28685075-3 2017 The aim of the present study was to evaluate the efficacy and adverse events (AE) of combination therapy with irinotecan and platinum (CPT-Pt) for ROC. Irinotecan 110-120 choline phosphotransferase 1 Homo sapiens 135-138 28025786-1 2017 Genetic variability in KRAS and EGFR predicts response to cetuximab in irinotecan refractory colorectal cancer. Irinotecan 71-81 KRAS proto-oncogene, GTPase Homo sapiens 23-27 28025786-1 2017 Genetic variability in KRAS and EGFR predicts response to cetuximab in irinotecan refractory colorectal cancer. Irinotecan 71-81 epidermal growth factor receptor Homo sapiens 32-36 28069724-0 2017 Synthetic Lethality Exploitation by an Anti-Trop-2-SN-38 Antibody-Drug Conjugate, IMMU-132, Plus PARP Inhibitors in BRCA1/2-wild-type Triple-Negative Breast Cancer. Irinotecan 51-56 tropomyosin 2, beta Mus musculus 44-50 28291390-2 2017 Sacituzumab govitecan, an antibody-drug conjugate, targets Trop-2 for the selective delivery of SN-38, the active metabolite of irinotecan. Irinotecan 96-101 tumor associated calcium signal transducer 2 Homo sapiens 59-65 28291390-2 2017 Sacituzumab govitecan, an antibody-drug conjugate, targets Trop-2 for the selective delivery of SN-38, the active metabolite of irinotecan. Irinotecan 128-138 tumor associated calcium signal transducer 2 Homo sapiens 59-65 28804517-8 2017 It is imperative to check UGT1A1 gene status in all patients being considered for treatment with nanoliposomal irinotecan. Irinotecan 111-121 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 26-32 28968978-0 2017 Decoy receptor 1 (DCR1) promoter hypermethylation and response to irinotecan in metastatic colorectal cancer. Irinotecan 66-76 TNF receptor superfamily member 10c Homo sapiens 0-16 28968978-0 2017 Decoy receptor 1 (DCR1) promoter hypermethylation and response to irinotecan in metastatic colorectal cancer. Irinotecan 66-76 TNF receptor superfamily member 10c Homo sapiens 18-22 28968978-7 2017 In conclusion, DCR1 promoter hypermethylation status is a potential predictive biomarker for response to treatment with irinotecan, when combined with capecitabine. Irinotecan 120-130 TNF receptor superfamily member 10c Homo sapiens 15-19 28157715-0 2017 Combined use of irinotecan with histone deacetylase inhibitor belinostat could cause severe toxicity by inhibiting SN-38 glucuronidation via UGT1A1. Irinotecan 16-26 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 141-147 28157715-3 2017 This inhibitory effect was determined by measuring the formation rate of SN-38 glucuronide after SN-38 incubation with human recombinant UGT1A isoforms (1A1, 1A6, 1A7 and 1A9) and pooled human liver microsomes (HLM, wild type, UGT1A1*1*28 and UGT1A1*28*28 allelic variants), with and without HDACis. Irinotecan 73-78 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 137-142 28157715-4 2017 The data showed that belinostat at 100 and 200 micromol/L inhibited SN-38 glucuronidation via UGT1A1 in a dose-dependent manner, causing significant decrease in Vmax and CLint (p < 0.05) from 12.60 to 1.95 pmol/min/mg and 21.59 to 4.20 muL/min/mg protein respectively. Irinotecan 68-73 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 94-100 28637434-1 2017 BACKGROUND: To evaluate a new UGT1A and DPYD polymorphism panel to better predict irinotecan-induced toxicity and the clinical response in Chinese patients with metastatic colorectal cancer (mCRC). Irinotecan 82-92 dihydropyrimidine dehydrogenase Homo sapiens 40-44 28637434-11 2017 In the first-line irinotecan-based treatment, UGT1A1*28 and DPYD*5 contributed to higher response rates (P = 0.043 and P = 0.019, respectively), while DPYD*5 was found to associate with better progression-free survival (P = 0.015). Irinotecan 18-28 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 46-52 28637434-11 2017 In the first-line irinotecan-based treatment, UGT1A1*28 and DPYD*5 contributed to higher response rates (P = 0.043 and P = 0.019, respectively), while DPYD*5 was found to associate with better progression-free survival (P = 0.015). Irinotecan 18-28 dihydropyrimidine dehydrogenase Homo sapiens 60-66 28637434-11 2017 In the first-line irinotecan-based treatment, UGT1A1*28 and DPYD*5 contributed to higher response rates (P = 0.043 and P = 0.019, respectively), while DPYD*5 was found to associate with better progression-free survival (P = 0.015). Irinotecan 18-28 dihydropyrimidine dehydrogenase Homo sapiens 151-157 28637434-13 2017 CONCLUSION: Results still showed UGT1A1*6 and UGT1A1*28 to be partially associated with irinotecan-induced toxicity and clinical response. Irinotecan 88-98 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-39 28637434-13 2017 CONCLUSION: Results still showed UGT1A1*6 and UGT1A1*28 to be partially associated with irinotecan-induced toxicity and clinical response. Irinotecan 88-98 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 46-52 28502040-0 2017 UGT1A1 polymorphisms with irinotecan-induced toxicities and treatment outcome in Asians with Lung Cancer: a meta-analysis. Irinotecan 26-36 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 28790841-0 2017 Relationship between UGT1A1*6/*28 gene polymorphisms and the efficacy and toxicity of irinotecan-based chemotherapy. Irinotecan 86-96 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 21-27 28790841-1 2017 PURPOSE: A retrospective study was performed to analyze the relationship between uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) *6/*28 gene polymorphisms and adverse reactions associated with irinotecan (CPT-11)-based chemotherapy. Irinotecan 202-212 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 81-128 28790841-1 2017 PURPOSE: A retrospective study was performed to analyze the relationship between uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) *6/*28 gene polymorphisms and adverse reactions associated with irinotecan (CPT-11)-based chemotherapy. Irinotecan 202-212 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 130-136 28790841-1 2017 PURPOSE: A retrospective study was performed to analyze the relationship between uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) *6/*28 gene polymorphisms and adverse reactions associated with irinotecan (CPT-11)-based chemotherapy. Irinotecan 214-220 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 81-128 28790841-2 2017 The correlation between UGT1A1 polymorphisms and the clinical efficacy of CPT-11 was also analyzed, along with the influence of age and tumor type. Irinotecan 74-80 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 24-30 28790841-5 2017 RESULTS: Colorectal cancer patients with the UGT1A1*6 mutant genotype had a significantly higher risk of severe delayed diarrhea than that of wild-type individuals when administered a CPT-11 dose >=130 mg/m2 (P=0.042); the same phenomenon was observed when the UGT1A1*6 and UGT1A1*28 mutant genotypes were considered together (P=0.028). Irinotecan 184-190 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 45-51 28790841-5 2017 RESULTS: Colorectal cancer patients with the UGT1A1*6 mutant genotype had a significantly higher risk of severe delayed diarrhea than that of wild-type individuals when administered a CPT-11 dose >=130 mg/m2 (P=0.042); the same phenomenon was observed when the UGT1A1*6 and UGT1A1*28 mutant genotypes were considered together (P=0.028). Irinotecan 184-190 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 264-270 28790841-5 2017 RESULTS: Colorectal cancer patients with the UGT1A1*6 mutant genotype had a significantly higher risk of severe delayed diarrhea than that of wild-type individuals when administered a CPT-11 dose >=130 mg/m2 (P=0.042); the same phenomenon was observed when the UGT1A1*6 and UGT1A1*28 mutant genotypes were considered together (P=0.028). Irinotecan 184-190 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 264-270 28790841-6 2017 However, in lung cancer patients administered a low dose of CPT-11, UGT1A1*6/*28 variants were not significantly associated with severe neutropenia or delayed diarrhea. Irinotecan 60-66 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 68-74 28586388-10 2017 Two had partial responses by RECIST 1.1 on a clinical trial involving a PD-1 inhibitor + irinotecan (indicated by MLH1 alteration). Irinotecan 89-99 mutL homolog 1 Homo sapiens 114-118 28502040-1 2017 Previous studies of irinotecan pharmacogenetics have shown that the UGT1A1*28 polymorphism has an effect on irinotecan (IRI)-induced toxicities in Caucasians. Irinotecan 20-30 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 68-74 28502040-1 2017 Previous studies of irinotecan pharmacogenetics have shown that the UGT1A1*28 polymorphism has an effect on irinotecan (IRI)-induced toxicities in Caucasians. Irinotecan 108-118 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 68-74 28502040-1 2017 Previous studies of irinotecan pharmacogenetics have shown that the UGT1A1*28 polymorphism has an effect on irinotecan (IRI)-induced toxicities in Caucasians. Irinotecan 120-123 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 68-74 28695301-7 2017 The influence of the chemotherapy backbone on anti-EGFR mAb efficacy, evaluated with meta-regression, indicated a higher ORR with irinotecan-based versus oxaliplatin-based regimens, but comparable PFS and OS. Irinotecan 130-140 epidermal growth factor receptor Homo sapiens 51-55 28439659-3 2017 This report demonstrated the outcomes of adjuvant nimotuzumab, an EGFR inhibitor, with irinotecan in pediatric HGGs. Irinotecan 87-97 epidermal growth factor receptor Homo sapiens 66-70 28081962-1 2017 BACKGROUND: Adding cetuximab to FOLFIRI (5-fluorouracil, leucovorin, irinotecan) significantly improved progression-free survival (PFS), overall survival (OS), and objective response rate (ORR) in patients with KRAS or RAS (KRAS/NRAS, exons 2-4) wild-type (wt) metastatic colorectal cancer (mCRC) in the first-line CRYSTAL study. Irinotecan 69-79 KRAS proto-oncogene, GTPase Homo sapiens 211-215 28081962-1 2017 BACKGROUND: Adding cetuximab to FOLFIRI (5-fluorouracil, leucovorin, irinotecan) significantly improved progression-free survival (PFS), overall survival (OS), and objective response rate (ORR) in patients with KRAS or RAS (KRAS/NRAS, exons 2-4) wild-type (wt) metastatic colorectal cancer (mCRC) in the first-line CRYSTAL study. Irinotecan 69-79 KRAS proto-oncogene, GTPase Homo sapiens 224-228 28081962-1 2017 BACKGROUND: Adding cetuximab to FOLFIRI (5-fluorouracil, leucovorin, irinotecan) significantly improved progression-free survival (PFS), overall survival (OS), and objective response rate (ORR) in patients with KRAS or RAS (KRAS/NRAS, exons 2-4) wild-type (wt) metastatic colorectal cancer (mCRC) in the first-line CRYSTAL study. Irinotecan 69-79 NRAS proto-oncogene, GTPase Homo sapiens 229-233 28242239-0 2017 Role of Toll-like receptor 4 in drug-drug interaction between paclitaxel and irinotecan in vitro. Irinotecan 77-87 toll like receptor 4 Homo sapiens 8-28 28242239-8 2017 Paclitaxel treatment increased the levels of irinotecan metabolites, SN-38 and SN-38 glucuronide (SN-38G) in TLR4-dependent manner. Irinotecan 45-55 toll like receptor 4 Homo sapiens 109-113 28242239-9 2017 Paclitaxel-mediated induction of genes involved in irinotecan metabolism such as Cyp3a11 and Ugt1a1 was TLR4-dependent, while induction of the transporter Mrp2 was TLR4-independent. Irinotecan 51-61 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-99 28242239-9 2017 Paclitaxel-mediated induction of genes involved in irinotecan metabolism such as Cyp3a11 and Ugt1a1 was TLR4-dependent, while induction of the transporter Mrp2 was TLR4-independent. Irinotecan 51-61 toll like receptor 4 Homo sapiens 104-108 28340475-0 2017 Irinotecan Upregulates Fibroblast Growth Factor Receptor 3 Expression in Colorectal Cancer Cells, Which Mitigates Irinotecan-Induced Apoptosis. Irinotecan 0-10 fibroblast growth factor receptor 3 Homo sapiens 23-58 28242239-8 2017 Paclitaxel treatment increased the levels of irinotecan metabolites, SN-38 and SN-38 glucuronide (SN-38G) in TLR4-dependent manner. Irinotecan 79-84 toll like receptor 4 Homo sapiens 109-113 28242239-10 2017 These novel findings demonstrate that paclitaxel can affect irinotecan metabolism by a TLR4-dependent mechanism. Irinotecan 60-70 toll like receptor 4 Homo sapiens 87-91 27938508-0 2017 Regorafenib Plus FOLFIRI With Irinotecan Dose Escalated According to Uridine Diphosphate Glucuronosyltransferase 1A1 Genotyping in Patients With Metastatic Colorectal Cancer. Irinotecan 30-40 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 69-116 28279925-0 2017 Irinotecan binds to the internal cavity of beta-lactoglobulin: A multi-spectroscopic and computational investigation. Irinotecan 0-10 beta-lactoglobulin Bos taurus 43-61 28279925-3 2017 Molecular docking results were in full agreement with the results obtained from thermodynamic analysis of the fluorescence data indicating the existence of one binding site for irinotecan in beta-LG structure and revealed the hydrophobic nature of the interaction between irinotecan and the protein. Irinotecan 177-187 beta-lactoglobulin Bos taurus 191-198 28279925-3 2017 Molecular docking results were in full agreement with the results obtained from thermodynamic analysis of the fluorescence data indicating the existence of one binding site for irinotecan in beta-LG structure and revealed the hydrophobic nature of the interaction between irinotecan and the protein. Irinotecan 272-282 beta-lactoglobulin Bos taurus 191-198 28279925-4 2017 The binding distance between beta-LG and irinotecan, r, was estimated to be 5.74nm based on the Forster"s theory of non-radiative energy transfer. Irinotecan 41-51 beta-lactoglobulin Bos taurus 29-36 28279925-6 2017 Based on the experimental data and the possible binding mode revealed by molecular docking study, we concluded that irinotecan binds to the hydrophobic calyx of beta-LG with induction of some alterations in the secondary and tertiary structure of the protein. Irinotecan 116-126 beta-lactoglobulin Bos taurus 161-168 27938508-8 2017 Our findings indicate that regorafenib plus FOLFIRI with irinotecan dose escalation based on UGT1A1 genotyping in previously treated patients with mCRC and with UGT1A1*1/*1 and UGT1A1*1/*28 genotypes is clinically effective and yields improved oncological outcomes. Irinotecan 57-67 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-99 28442401-9 2017 In sum, our findings indicate that TRIB3 plays an anti-apoptotic role in doxorubicin-treated gastric cancer cell lines, perhaps indicating that the status of TRIB3 expression in response to anticancer drugs, such as doxorubicin, irinotecan or oxaliplatin, may reflect the efficiency for cancer therapy. Irinotecan 229-239 tribbles pseudokinase 3 Homo sapiens 35-40 28596680-12 2017 AXL silencing showed a subtle trend to restore colon cancer cell sensitivity to 5FU or irinotecan. Irinotecan 87-97 AXL receptor tyrosine kinase Homo sapiens 0-3 28442401-9 2017 In sum, our findings indicate that TRIB3 plays an anti-apoptotic role in doxorubicin-treated gastric cancer cell lines, perhaps indicating that the status of TRIB3 expression in response to anticancer drugs, such as doxorubicin, irinotecan or oxaliplatin, may reflect the efficiency for cancer therapy. Irinotecan 229-239 tribbles pseudokinase 3 Homo sapiens 158-163 27641154-7 2017 Polymorphisms in DPYD, TPMT, and UGT1A1 have been described that have a major impact on the pharmacokinetics of 5-fluorouracil, mercaptopurine, and irinotecan, respectively. Irinotecan 148-158 dihydropyrimidine dehydrogenase Homo sapiens 17-21 28468698-8 2017 The proposed method was successfully applied to the determination of irinotecan in tap water, river water, and urine samples spiked with 10.20 mg/L for the water samples and 8.32 mg/L for the urine sample. Irinotecan 69-79 nuclear RNA export factor 1 Homo sapiens 83-86 28468698-9 2017 The average recovery values of irinotecan determined were 99.1% for tap water, 109.4% for river water, and 96.1% for urine. Irinotecan 31-41 nuclear RNA export factor 1 Homo sapiens 68-71 28414297-4 2017 In a KRAS-induced orthotopic PDAC model, coadministration of iRGD enhanced the uptake of an irinotecan-loaded silicasome carrier that comprises lipid bilayer-coated mesoporous silica nanoparticles (MSNPs); this uptake resulted in enhanced survival and markedly reduced metastasis. Irinotecan 92-102 KRAS proto-oncogene, GTPase Homo sapiens 5-9 28265824-0 2017 Relationship between magnetic resonance imaging characteristics and plasmatic levels of MMP2 and MMP9 in patients with recurrent high-grade gliomas treated by Bevacizumab and Irinotecan. Irinotecan 175-185 matrix metallopeptidase 2 Homo sapiens 88-92 28265824-0 2017 Relationship between magnetic resonance imaging characteristics and plasmatic levels of MMP2 and MMP9 in patients with recurrent high-grade gliomas treated by Bevacizumab and Irinotecan. Irinotecan 175-185 matrix metallopeptidase 9 Homo sapiens 97-101 28267710-0 2017 Dasatinib synergises with irinotecan to suppress hepatocellular carcinoma via inhibiting the protein synthesis of PLK1. Irinotecan 26-36 polo like kinase 1 Homo sapiens 114-118 28267710-14 2017 CONCLUSIONS: Dasatinib is able to reinforce the anti-HCC efficacy of irinotecan/SN38 by downregulation of PLK1 synthesis. Irinotecan 69-79 polo like kinase 1 Homo sapiens 106-110 28388962-9 2017 Concomitantly, CPT-11 enhanced tumor necrosis factor-alpha (TNF-alpha) and interleukin-6 (IL-6) levels and inducible nitric oxide synthase (iNOS) gene expression, associated with an increase in the total number macrophages (positive cells for ionized calcium-binding adapter molecule, Iba-1) and degranulated mast cells in the small intestine segments and caused significant weight loss. Irinotecan 15-21 tumor necrosis factor Mus musculus 60-69 28388962-9 2017 Concomitantly, CPT-11 enhanced tumor necrosis factor-alpha (TNF-alpha) and interleukin-6 (IL-6) levels and inducible nitric oxide synthase (iNOS) gene expression, associated with an increase in the total number macrophages (positive cells for ionized calcium-binding adapter molecule, Iba-1) and degranulated mast cells in the small intestine segments and caused significant weight loss. Irinotecan 15-21 interleukin 6 Mus musculus 75-88 26245851-0 2017 Phase II study of cetuximab with irinotecan for KRAS wild-type colorectal cancer in Japanese patients. Irinotecan 33-43 KRAS proto-oncogene, GTPase Homo sapiens 48-52 28288854-0 2017 Irinotecan-induced bile acid malabsorption is associated with down-regulation of ileal Asbt (Slc10a2) in mice. Irinotecan 0-10 solute carrier family 10, member 2 Mus musculus 93-100 28220486-1 2017 The addition of irinotecan to an epidermal growth factor receptor (EGFR) antibody has previously been shown to improve tumor response rate and time to progression but not overall survival (OS) for refractory metastatic colorectal cancer (mCRC). Irinotecan 16-26 epidermal growth factor receptor Homo sapiens 33-65 28220486-1 2017 The addition of irinotecan to an epidermal growth factor receptor (EGFR) antibody has previously been shown to improve tumor response rate and time to progression but not overall survival (OS) for refractory metastatic colorectal cancer (mCRC). Irinotecan 16-26 epidermal growth factor receptor Homo sapiens 67-71 28131876-5 2017 Cabozantinib, at non-toxic concentrations (3 or 5muM), sensitized the ABCG2-overexpressing cells to mitoxantrone, SN-38, and topotecan. Irinotecan 114-119 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 70-75 28388962-9 2017 Concomitantly, CPT-11 enhanced tumor necrosis factor-alpha (TNF-alpha) and interleukin-6 (IL-6) levels and inducible nitric oxide synthase (iNOS) gene expression, associated with an increase in the total number macrophages (positive cells for ionized calcium-binding adapter molecule, Iba-1) and degranulated mast cells in the small intestine segments and caused significant weight loss. Irinotecan 15-21 interleukin 6 Mus musculus 90-94 28388962-9 2017 Concomitantly, CPT-11 enhanced tumor necrosis factor-alpha (TNF-alpha) and interleukin-6 (IL-6) levels and inducible nitric oxide synthase (iNOS) gene expression, associated with an increase in the total number macrophages (positive cells for ionized calcium-binding adapter molecule, Iba-1) and degranulated mast cells in the small intestine segments and caused significant weight loss. Irinotecan 15-21 nitric oxide synthase 2, inducible Mus musculus 107-138 28388962-9 2017 Concomitantly, CPT-11 enhanced tumor necrosis factor-alpha (TNF-alpha) and interleukin-6 (IL-6) levels and inducible nitric oxide synthase (iNOS) gene expression, associated with an increase in the total number macrophages (positive cells for ionized calcium-binding adapter molecule, Iba-1) and degranulated mast cells in the small intestine segments and caused significant weight loss. Irinotecan 15-21 nitric oxide synthase 2, inducible Mus musculus 140-144 28388962-9 2017 Concomitantly, CPT-11 enhanced tumor necrosis factor-alpha (TNF-alpha) and interleukin-6 (IL-6) levels and inducible nitric oxide synthase (iNOS) gene expression, associated with an increase in the total number macrophages (positive cells for ionized calcium-binding adapter molecule, Iba-1) and degranulated mast cells in the small intestine segments and caused significant weight loss. Irinotecan 15-21 induction of brown adipocytes 1 Mus musculus 285-290 27834032-9 2017 CD107a+/ROSlow cells were enriched in HT29 and DLD1 cultures after treatments with oxaliplatin, 5-fluorouracil, and the irinotecan metabolite SN38. Irinotecan 120-130 lysosomal associated membrane protein 1 Homo sapiens 0-6 28314987-0 2017 Cooperative inhibitory effects of uremic toxins and other serum components on OATP1B1-mediated transport of SN-38. Irinotecan 108-113 solute carrier organic anion transporter family member 1B1 Homo sapiens 78-85 28314987-2 2017 Uremic toxins (UTs), which accumulate in the serum of ESKD patients, were reported to inhibit organic anion-transporting polypeptide (OATP) 1B1-mediated uptake of SN-38; however, the relevance of this finding in a clinical setting is unknown. Irinotecan 163-168 solute carrier organic anion transporter family member 1B1 Homo sapiens 94-143 28314987-3 2017 This study focused on cooperative effects of serum components and UTs on OATP1B1-mediated transport of SN-38. Irinotecan 103-108 solute carrier organic anion transporter family member 1B1 Homo sapiens 73-80 28314987-4 2017 METHODS: Uptake of SN-38 by OATP1B1 was evaluated using cells stably expressing OATP1B1. Irinotecan 19-24 solute carrier organic anion transporter family member 1B1 Homo sapiens 28-35 28314987-4 2017 METHODS: Uptake of SN-38 by OATP1B1 was evaluated using cells stably expressing OATP1B1. Irinotecan 19-24 solute carrier organic anion transporter family member 1B1 Homo sapiens 80-87 28314987-7 2017 RESULTS: Uptake clearance of SN-38 in OATP1B1 cells decreased by 40% in the presence of uremic serum residue with albumin compared to that in the presence of normal serum residue. Irinotecan 29-34 solute carrier organic anion transporter family member 1B1 Homo sapiens 38-45 28314987-10 2017 CONCLUSIONS: Hepatic uptake of SN-38 via OATP1B1 decreases in ESKD patients through cooperative inhibitory effects of UTs and serum components. Irinotecan 31-36 solute carrier organic anion transporter family member 1B1 Homo sapiens 41-48 27641154-7 2017 Polymorphisms in DPYD, TPMT, and UGT1A1 have been described that have a major impact on the pharmacokinetics of 5-fluorouracil, mercaptopurine, and irinotecan, respectively. Irinotecan 148-158 thiopurine S-methyltransferase Homo sapiens 23-27 27641154-7 2017 Polymorphisms in DPYD, TPMT, and UGT1A1 have been described that have a major impact on the pharmacokinetics of 5-fluorouracil, mercaptopurine, and irinotecan, respectively. Irinotecan 148-158 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-39 28443467-3 2017 In this study, the effect of irinotecan on high-mobility group protein B1 and MMP9 content, gene expression, cell cycle, and cell growth in human breast cancer cells (MCF-7) was investigated. Irinotecan 29-39 high mobility group box 1 Homo sapiens 43-73 28087739-6 2017 Downregulation of RIP1 in a xenograft model impaired tumor growth inhibition from SN38 treatment, suggesting the potential of RIP1 to determine the clinical outcome of irinotecan treatment. Irinotecan 168-178 receptor interacting serine/threonine kinase 1 Homo sapiens 18-22 28262261-3 2017 This review presents the usefulness of pharmacogenetic tests for some key applications: dihydropyrimidine dehydrogenase (DPYD) genotyping for fluoropyrimidine (5-fluorouracil, capecitabine), UDP glucuronosylstransferase (UGT1A1) for irinotecan and thiopurine S-methyltransferase (TPMT) for thiopurine drugs. Irinotecan 233-243 dihydropyrimidine dehydrogenase Homo sapiens 88-119 28262261-3 2017 This review presents the usefulness of pharmacogenetic tests for some key applications: dihydropyrimidine dehydrogenase (DPYD) genotyping for fluoropyrimidine (5-fluorouracil, capecitabine), UDP glucuronosylstransferase (UGT1A1) for irinotecan and thiopurine S-methyltransferase (TPMT) for thiopurine drugs. Irinotecan 233-243 dihydropyrimidine dehydrogenase Homo sapiens 121-125 28443467-3 2017 In this study, the effect of irinotecan on high-mobility group protein B1 and MMP9 content, gene expression, cell cycle, and cell growth in human breast cancer cells (MCF-7) was investigated. Irinotecan 29-39 matrix metallopeptidase 9 Homo sapiens 78-82 28443467-9 2017 Irinotecan decreased H3K9 acetylation and increased poly ADP-ribose polymerase fragmentation to 89 kDa and anion superoxide production suggesting induction of apoptosis in these cells. Irinotecan 0-10 poly(ADP-ribose) polymerase 1 Homo sapiens 52-78 28443467-11 2017 From the results, it is concluded that overexpression of high-mobility group protein B1 in the presence of irinotecan precedes breast cancer cells into apoptosis and in this response the binding of irinotecan to chromatin or high-mobility group protein B1 may condense/aggregate chromatin, preventing high-mobility group protein B1 release from chromatin. Irinotecan 107-117 high mobility group box 1 Homo sapiens 57-87 28443467-11 2017 From the results, it is concluded that overexpression of high-mobility group protein B1 in the presence of irinotecan precedes breast cancer cells into apoptosis and in this response the binding of irinotecan to chromatin or high-mobility group protein B1 may condense/aggregate chromatin, preventing high-mobility group protein B1 release from chromatin. Irinotecan 198-208 high mobility group box 1 Homo sapiens 57-87 28278081-0 2017 Correlative analysis of plasma SN-38 levels and DPD activity with outcomes of FOLFIRI regimen for metastatic colorectal cancer with UGT1A1 *28 and *6 wild type and its implication for individualized chemotherapy. Irinotecan 31-36 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 132-138 29069732-5 2017 Secondly, based on the concept of a bi-therapy targeting in pHGGs mTORC1 (rapamycin) and HIF-1alpha (irinotecan), we hypothesized that the balanced expressions between RAS/ERK, PI3K/AKT and HIF-1alpha/HIF-2alpha/MYC proteins or genes may provide a modulation of the cell response to this double targeting. Irinotecan 101-111 hypoxia inducible factor 1 subunit alpha Homo sapiens 89-99 28278081-1 2017 It remains uncertain whether there is an correlation between clinical pharmacokinetic parameters and outcomes for metastatic colorectal cancer especially with UGT1A1 *28 and *6 wild type (*1/*1-*1/*1) for serious events associated with Irinotecan are largely excluded. Irinotecan 236-246 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 159-165 28278081-2 2017 This study retrospectively analyzed the relationship between Irinotecan metabolite levels and outcomes of UGT1A1 *1/*1-*1/*1 genotype arrangement. Irinotecan 61-71 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 106-112 28278081-8 2017 Therefore, plasma SN-38 levels is related to outcomes for UGT1A1 *1/*1-*1/*1 genotype, to improve efficacy, patients with CSN-38 1.5 h lower than 50.24 ng/ml, CPT-11 dosage could be added in next chemmotherapy on SN-38 plasma level monitoring. Irinotecan 18-23 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 58-64 28255900-9 2017 The differences between HSPC1 inhibitors were also reflected in combination treatment-17-DMAG was more effective compared with NVP-AUY922 in potentiating the cytotoxic effects of 5-fluorouracil, oxaliplatin and irinotecan. Irinotecan 211-221 heat shock protein 90 alpha family class A member 1 Homo sapiens 24-29 28000054-11 2017 Furthermore, 5-FU retains its ability to cause nuclear accumulation of p53 in the presence of irinotecan or topotecan. Irinotecan 94-104 tumor protein p53 Homo sapiens 71-74 28280378-0 2017 UGT1A1 polymorphisms in cancer: impact on irinotecan treatment. Irinotecan 42-52 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 28126414-3 2017 hCE1 is known to play crucial roles in the metabolism of a wide variety of endogenous esters, clinical drugs and insecticides, while hCE2 plays a key role in the metabolic activation of anticancer agents including irinotecan and capecitabine. Irinotecan 214-224 carboxylesterase 2 Homo sapiens 133-137 28131902-0 2017 CQ synergistically sensitizes human colorectal cancer cells to SN-38/CPT-11 through lysosomal and mitochondrial apoptotic pathway via p53-ROS cross-talk. Irinotecan 63-68 tumor protein p53 Homo sapiens 134-137 28131902-0 2017 CQ synergistically sensitizes human colorectal cancer cells to SN-38/CPT-11 through lysosomal and mitochondrial apoptotic pathway via p53-ROS cross-talk. Irinotecan 69-75 tumor protein p53 Homo sapiens 134-137 28131902-4 2017 Notably, another autophagy inhibitor chloroquine (CQ) synergistically enhanced the anti-tumor activity of SN-38 in CRC cells with wild type (WT) p53. Irinotecan 106-111 tumor protein p53 Homo sapiens 145-148 28131902-5 2017 Subsequently, we identified a potential mechanism of this cooperative interaction by showing that CQ and SN-38 acted together to trigger reactive oxygen species (ROS) burst, upregulate p53 expression, elicit the loss of lysosomal membrane potential (LMP) and mitochondrial membrane potential ( psim). Irinotecan 105-110 tumor protein p53 Homo sapiens 185-188 28131902-6 2017 In addition, ROS induced by CQ plus SN-38 upregulated p53 levels by activating p38, conversely, p53 stimulated ROS. Irinotecan 36-41 tumor protein p53 Homo sapiens 54-57 28131902-6 2017 In addition, ROS induced by CQ plus SN-38 upregulated p53 levels by activating p38, conversely, p53 stimulated ROS. Irinotecan 36-41 mitogen-activated protein kinase 14 Homo sapiens 79-82 28131902-9 2017 Altogether, all results suggested that CQ synergistically sensitized human CRC cells with WT p53 to SN-38 through lysosomal and mitochondrial apoptotic pathway via p53-ROS cross-talk. Irinotecan 100-105 tumor protein p53 Homo sapiens 93-96 27169843-7 2017 In tumor specimens, cytoplasmic accumulation of BLM correlates with DNA damage and gammaH2AX and phosphorylated ATM foci and predicts long-term progression-free survival in metastatic patients treated with irinotecan. Irinotecan 206-216 BLM RecQ like helicase Homo sapiens 48-51 27169843-7 2017 In tumor specimens, cytoplasmic accumulation of BLM correlates with DNA damage and gammaH2AX and phosphorylated ATM foci and predicts long-term progression-free survival in metastatic patients treated with irinotecan. Irinotecan 206-216 ATM serine/threonine kinase Homo sapiens 112-115 28280378-3 2017 Among them, UGT1A1*28 and UGT1A1*6 have been reported to be associated with severe toxicities in patients treated with irinotecan-based chemotherapy by increasing the dose of SN-38 (7-ethyl-10-hydroxycamptothecin), an active form of irinotecan. Irinotecan 119-129 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-18 28280378-3 2017 Among them, UGT1A1*28 and UGT1A1*6 have been reported to be associated with severe toxicities in patients treated with irinotecan-based chemotherapy by increasing the dose of SN-38 (7-ethyl-10-hydroxycamptothecin), an active form of irinotecan. Irinotecan 119-129 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 26-32 28280378-3 2017 Among them, UGT1A1*28 and UGT1A1*6 have been reported to be associated with severe toxicities in patients treated with irinotecan-based chemotherapy by increasing the dose of SN-38 (7-ethyl-10-hydroxycamptothecin), an active form of irinotecan. Irinotecan 175-180 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-18 28280378-3 2017 Among them, UGT1A1*28 and UGT1A1*6 have been reported to be associated with severe toxicities in patients treated with irinotecan-based chemotherapy by increasing the dose of SN-38 (7-ethyl-10-hydroxycamptothecin), an active form of irinotecan. Irinotecan 175-180 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 26-32 28280378-3 2017 Among them, UGT1A1*28 and UGT1A1*6 have been reported to be associated with severe toxicities in patients treated with irinotecan-based chemotherapy by increasing the dose of SN-38 (7-ethyl-10-hydroxycamptothecin), an active form of irinotecan. Irinotecan 182-212 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-18 28280378-3 2017 Among them, UGT1A1*28 and UGT1A1*6 have been reported to be associated with severe toxicities in patients treated with irinotecan-based chemotherapy by increasing the dose of SN-38 (7-ethyl-10-hydroxycamptothecin), an active form of irinotecan. Irinotecan 182-212 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 26-32 28280378-3 2017 Among them, UGT1A1*28 and UGT1A1*6 have been reported to be associated with severe toxicities in patients treated with irinotecan-based chemotherapy by increasing the dose of SN-38 (7-ethyl-10-hydroxycamptothecin), an active form of irinotecan. Irinotecan 233-243 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-18 28280378-3 2017 Among them, UGT1A1*28 and UGT1A1*6 have been reported to be associated with severe toxicities in patients treated with irinotecan-based chemotherapy by increasing the dose of SN-38 (7-ethyl-10-hydroxycamptothecin), an active form of irinotecan. Irinotecan 233-243 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 26-32 28280378-4 2017 Many association studies and meta-analyses have demonstrated the contribution of UGT1A1*28 and UGT1A1*6 polymorphisms to the toxicities caused by irinotecan-based therapy. Irinotecan 146-156 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 81-87 28367249-0 2017 Predictive Value of UGT1A1*28 Polymorphism In Irinotecan-based Chemotherapy. Irinotecan 46-56 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 20-26 28367249-1 2017 The UGT1A1*28 polymorphism was suggested to be significantly connected with irinotecan-induced toxicity and response to chemotherapy. Irinotecan 76-86 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 28280378-4 2017 Many association studies and meta-analyses have demonstrated the contribution of UGT1A1*28 and UGT1A1*6 polymorphisms to the toxicities caused by irinotecan-based therapy. Irinotecan 146-156 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 95-101 28367249-3 2017 Hence we carried out a meta-analysis to investigate the effect of UGT1A1*28 polymorphism on severe diarrhea, neutropenia, and response of patients who had undergone irinotecan-based chemotherapy. Irinotecan 165-175 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 66-72 28173214-0 2017 UGT1A1 polymorphism as a prognostic indicator of stage I ovarian clear cell carcinoma patients treated with irinotecan. Irinotecan 108-118 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 28367249-10 2017 Our data showed that the UGT1A1*28 polymorphism had a significant relationship with toxicity and response to irinotecan-based chemotherapy. Irinotecan 109-119 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 25-31 28112927-1 2017 Carboxylesterases (CEs) are ubiquitous enzymes that are responsible for the metabolism of xenobiotics, including drugs such as irinotecan and oseltamivir. Irinotecan 127-137 carboxylesterase 1 Homo sapiens 0-17 28180300-5 2017 Subsequent treatment of irinotecan-resistant, but not parental, CRC cells with histone deacetylase (HDAC) inhibitors can effectively overcome resistance. Irinotecan 24-34 histone deacetylase 9 Homo sapiens 79-98 28180300-5 2017 Subsequent treatment of irinotecan-resistant, but not parental, CRC cells with histone deacetylase (HDAC) inhibitors can effectively overcome resistance. Irinotecan 24-34 histone deacetylase 9 Homo sapiens 100-104 28180300-8 2017 These findings identify perturbed chromatin acetylation in irinotecan resistance and establish HDAC inhibitors as potential therapeutic means to overcome resistance. Irinotecan 59-69 histone deacetylase 9 Homo sapiens 95-99 27507617-1 2017 Purpose:UGT1A1*28 confers a higher risk of toxicity in patients treated with irinotecan. Irinotecan 77-87 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 8-14 27507617-3 2017 We aimed to identify the MTD of irinotecan in patients with metastatic colorectal cancer (mCRC) with *1/*1 and *1/*28 genotypes treated with FOLFIRI plus bevacizumab, and to determine whether bevacizumab alters irinotecan pharmacokinetics.Experimental Design: Previously untreated patients with mCRC (25 *1/*1; 23 *1/*28) were given FOLFIRI plus bevacizumab every 2 weeks. Irinotecan 32-42 metallothionein 1E Homo sapiens 25-28 27507617-3 2017 We aimed to identify the MTD of irinotecan in patients with metastatic colorectal cancer (mCRC) with *1/*1 and *1/*28 genotypes treated with FOLFIRI plus bevacizumab, and to determine whether bevacizumab alters irinotecan pharmacokinetics.Experimental Design: Previously untreated patients with mCRC (25 *1/*1; 23 *1/*28) were given FOLFIRI plus bevacizumab every 2 weeks. Irinotecan 211-221 metallothionein 1E Homo sapiens 25-28 27507617-4 2017 The irinotecan dose was escalated using a 3 + 3 design in each genotype group as follows: 260, 310, and 370 mg/m2 The MTD was the highest dose at which <4/10 patients had a dose-limiting toxicity (DLT). Irinotecan 4-14 metallothionein 1E Homo sapiens 118-121 27507617-8 2017 Changes in the AUCs of irinotecan and SN-38 associated with bevacizumab treatment were marginal.Conclusions: The MTD of irinotecan in FOLFIRI plus bevacizumab is 310 mg/m2 for UGT1A1 *1/*1 patients and 260 mg/m2 for *1/*28 patients. Irinotecan 120-130 metallothionein 1E Homo sapiens 113-116 27507617-8 2017 Changes in the AUCs of irinotecan and SN-38 associated with bevacizumab treatment were marginal.Conclusions: The MTD of irinotecan in FOLFIRI plus bevacizumab is 310 mg/m2 for UGT1A1 *1/*1 patients and 260 mg/m2 for *1/*28 patients. Irinotecan 120-130 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 176-182 28052029-8 2017 Moreover, RLYE showed a synergistic effect of the cytotoxic agent irinotecan on tumor cell apoptosis and tumor progression via tumor vessel normalization due to stabilization of VE-cadherin-mediated adherens junction, improvement of pericyte coverage, and inhibition of vascular leakage in tumors. Irinotecan 66-76 cadherin 5 Mus musculus 178-189 28173214-1 2017 We investigated whether UGT1A1 polymorphisms are associated with the prognosis of ovarian cancer patients treated with irinotecan. Irinotecan 119-129 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 24-30 28173214-7 2017 We conclude that UGT1A1 polymorphisms have the potential to be a prognostic marker of irinotecan treatment. Irinotecan 86-96 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 17-23 27060453-0 2017 Inhibition of NF-kappaB and metastasis in irinotecan (CPT-11)-resistant LoVo colon cancer cells by thymoquinone via JNK and p38. Irinotecan 42-52 mitogen-activated protein kinase 1 Homo sapiens 124-127 28011980-0 2017 Potential safety concerns of TLR4 antagonism with irinotecan: a preclinical observational report. Irinotecan 50-60 toll-like receptor 4 Rattus norvegicus 29-33 28011980-1 2017 PURPOSE: Irinotecan-induced gut toxicity is mediated in part by Toll-Like receptor 4 (TLR4) signalling. Irinotecan 9-19 toll-like receptor 4 Rattus norvegicus 64-84 28011980-1 2017 PURPOSE: Irinotecan-induced gut toxicity is mediated in part by Toll-Like receptor 4 (TLR4) signalling. Irinotecan 9-19 toll-like receptor 4 Rattus norvegicus 86-90 28011980-2 2017 The primary purpose of this preclinical study was to determine whether blocking TLR4 signalling by administering (-)-naloxone, a TLR4 antagonist, would improve irinotecan-induced gut toxicity. Irinotecan 160-170 toll-like receptor 4 Rattus norvegicus 80-84 27770239-0 2017 Irinotecan-induced mucositis: the interactions and potential role of GLP-2 analogues. Irinotecan 0-10 glucagon Homo sapiens 69-74 27060453-0 2017 Inhibition of NF-kappaB and metastasis in irinotecan (CPT-11)-resistant LoVo colon cancer cells by thymoquinone via JNK and p38. Irinotecan 54-60 mitogen-activated protein kinase 1 Homo sapiens 124-127 27060453-5 2017 Irinotecan resistant (CPT-11-R) LoVo colon cancer cell line was previous constructed by step-wise CPT-11 challenges to un-treated parental LoVo cells and expresses EGFR/IKKalpha/beta/NF-kappaB pathway. Irinotecan 0-10 epidermal growth factor receptor Homo sapiens 164-168 27060453-5 2017 Irinotecan resistant (CPT-11-R) LoVo colon cancer cell line was previous constructed by step-wise CPT-11 challenges to un-treated parental LoVo cells and expresses EGFR/IKKalpha/beta/NF-kappaB pathway. Irinotecan 0-10 component of inhibitor of nuclear factor kappa B kinase complex Homo sapiens 169-182 27060453-5 2017 Irinotecan resistant (CPT-11-R) LoVo colon cancer cell line was previous constructed by step-wise CPT-11 challenges to un-treated parental LoVo cells and expresses EGFR/IKKalpha/beta/NF-kappaB pathway. Irinotecan 22-28 epidermal growth factor receptor Homo sapiens 164-168 27060453-5 2017 Irinotecan resistant (CPT-11-R) LoVo colon cancer cell line was previous constructed by step-wise CPT-11 challenges to un-treated parental LoVo cells and expresses EGFR/IKKalpha/beta/NF-kappaB pathway. Irinotecan 22-28 component of inhibitor of nuclear factor kappa B kinase complex Homo sapiens 169-182 29956901-11 2017 CONCLUSION: SXD could significantly reduce irinotecan-induced hematological and gastrointestinaltoxicities in UGT1A1*28 or *6 variant patients (high risk group), while this treatment didn"t affect clinicalresponse of chemotherapy. Irinotecan 43-53 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 110-116 27324368-7 2017 In vivo data were in agreement with Ki67 staining, showing a significantly lower percentage of Ki67-positive cells in the irinotecan group as compared with other groups (p <= 0.0001). Irinotecan 122-132 antigen identified by monoclonal antibody Ki 67 Mus musculus 36-40 27324368-7 2017 In vivo data were in agreement with Ki67 staining, showing a significantly lower percentage of Ki67-positive cells in the irinotecan group as compared with other groups (p <= 0.0001). Irinotecan 122-132 antigen identified by monoclonal antibody Ki 67 Mus musculus 95-99 27324368-8 2017 Tumor expression of thymidine kinase 1 phosphorylated on serine 13, thymidylate synthase, and thymidine phosphorylase remained unaffected, while thymidine kinase 1 expression was, surprisingly, significantly higher in irinotecan-treated animals (p <= 0.01). Irinotecan 218-228 thymidine kinase 1 Mus musculus 145-163 28395759-2 2017 Patients were stratified by their pharmacogenetic/phenotypic status: the irinotecan dose was adjusted according to the number of TA tandem repeats in the UGT1A1 promoter, while the 5-fluorouracil (5-FU) dose was initially adjusted according to dihydropyrimidine dehydrogenase (DPD) activity at initial screening (5-FUODPM Tox) followed by PK-guided dose optimization (5-FUODPM Protocol). Irinotecan 73-83 dihydropyrimidine dehydrogenase Homo sapiens 244-275 28395759-2 2017 Patients were stratified by their pharmacogenetic/phenotypic status: the irinotecan dose was adjusted according to the number of TA tandem repeats in the UGT1A1 promoter, while the 5-fluorouracil (5-FU) dose was initially adjusted according to dihydropyrimidine dehydrogenase (DPD) activity at initial screening (5-FUODPM Tox) followed by PK-guided dose optimization (5-FUODPM Protocol). Irinotecan 73-83 dihydropyrimidine dehydrogenase Homo sapiens 277-280 28395759-2 2017 Patients were stratified by their pharmacogenetic/phenotypic status: the irinotecan dose was adjusted according to the number of TA tandem repeats in the UGT1A1 promoter, while the 5-fluorouracil (5-FU) dose was initially adjusted according to dihydropyrimidine dehydrogenase (DPD) activity at initial screening (5-FUODPM Tox) followed by PK-guided dose optimization (5-FUODPM Protocol). Irinotecan 73-83 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 154-160 28395759-2 2017 Patients were stratified by their pharmacogenetic/phenotypic status: the irinotecan dose was adjusted according to the number of TA tandem repeats in the UGT1A1 promoter, while the 5-fluorouracil (5-FU) dose was initially adjusted according to dihydropyrimidine dehydrogenase (DPD) activity at initial screening (5-FUODPM Tox) followed by PK-guided dose optimization (5-FUODPM Protocol). Irinotecan 73-83 thymocyte selection associated high mobility group box Homo sapiens 322-325 28011495-8 2017 Granulocyte colony-stimulating factor (G-CSF) in serum was elevated at 5.6 pg/ml, which further raised to 43 pg/ml one week after FOLFIRINOX chemotherapy (oxaliplatin, irinotecan, 5-fluorouracil), while WBC decreased from 103.3 G/l to 59.3 G/l. Irinotecan 168-178 colony stimulating factor 3 Homo sapiens 0-37 27650103-7 2017 P-selectin expression decreased significantly in the jejunum following irinotecan. Irinotecan 71-81 selectin P Rattus norvegicus 0-10 27650103-10 2017 CONCLUSIONS: Irinotecan induced a significant alteration in CAM expression in the jejunum and colon. Irinotecan 13-23 calmodulin 1 Rattus norvegicus 60-63 27959571-9 2017 These RDT-coated nanoparticles (RDT-NP) were further used for preferential delivery of Irinotecan, a chemotherapeutic agent, to cells overexpressing HB-EGF. Irinotecan 87-97 SWI/SNF related, matrix associated, actin dependent regulator of chromatin, subfamily b, member 1 Homo sapiens 6-9 27959571-9 2017 These RDT-coated nanoparticles (RDT-NP) were further used for preferential delivery of Irinotecan, a chemotherapeutic agent, to cells overexpressing HB-EGF. Irinotecan 87-97 SWI/SNF related, matrix associated, actin dependent regulator of chromatin, subfamily b, member 1 Homo sapiens 32-35 27959571-10 2017 We show that receptor-mediated enhanced uptake of RDT-NPs increases the potency of irinotecan in these cells. Irinotecan 83-93 SWI/SNF related, matrix associated, actin dependent regulator of chromatin, subfamily b, member 1 Homo sapiens 50-53 28011495-8 2017 Granulocyte colony-stimulating factor (G-CSF) in serum was elevated at 5.6 pg/ml, which further raised to 43 pg/ml one week after FOLFIRINOX chemotherapy (oxaliplatin, irinotecan, 5-fluorouracil), while WBC decreased from 103.3 G/l to 59.3 G/l. Irinotecan 168-178 colony stimulating factor 3 Homo sapiens 39-44 29098099-0 2017 Life-Threatening Irinotecan-Induced Toxicity in an Adult Patient with Alveolar Rhabdomyosarcoma: The Role of a UGT1A1 Polymorphism. Irinotecan 17-27 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 111-117 29098099-7 2017 The analysis of the uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) gene revealed homozygous UGT1A1 *28 polymorphism with an associated homozygous mutation c.-3275T>G; the latter is associated with a decrease of about 80% of UGT1A1 transcription explaining this irinotecan induced toxicity. Irinotecan 274-284 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 20-67 29098099-7 2017 The analysis of the uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) gene revealed homozygous UGT1A1 *28 polymorphism with an associated homozygous mutation c.-3275T>G; the latter is associated with a decrease of about 80% of UGT1A1 transcription explaining this irinotecan induced toxicity. Irinotecan 274-284 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 69-75 29098099-7 2017 The analysis of the uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) gene revealed homozygous UGT1A1 *28 polymorphism with an associated homozygous mutation c.-3275T>G; the latter is associated with a decrease of about 80% of UGT1A1 transcription explaining this irinotecan induced toxicity. Irinotecan 274-284 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 102-108 29098099-7 2017 The analysis of the uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) gene revealed homozygous UGT1A1 *28 polymorphism with an associated homozygous mutation c.-3275T>G; the latter is associated with a decrease of about 80% of UGT1A1 transcription explaining this irinotecan induced toxicity. Irinotecan 274-284 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 102-108 29098099-8 2017 Physician must be aware of the potential hematological (mainly neutropenia and infectious disease) and digestive (mainly diarrhea) toxicities caused by irinotecan and especially when the patient presents a UGT1A1 *28 homozygous allele. Irinotecan 152-162 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 206-212 29098099-9 2017 UGT1A genotyping performed before initiating treatment is useful to anticipate severe toxic reaction to irinotecan and improve the benefit/risk ratio of its use. Irinotecan 104-114 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 0-5 27503581-5 2017 Both UGT1A1*6 and *28 were reliably demonstrated to be risk factors for irinotecan-induced neutropenia, with tests for both polymorphisms potentially being particularly useful in Asian cancer patients. Irinotecan 72-82 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 5-11 28167974-11 2017 Additionally, SXD decreased the activity of beta-glucuronidase after irinotecan administration. Irinotecan 69-79 glucuronidase, beta Rattus norvegicus 44-62 28881354-1 2017 OBJECTIVES: This phase II trial was aimed at assessing the safety and activity of capecitabine, oxaliplatin, and irinotecan (COI regimen) as a preoperative treatment for resectable gastric cancer (GC) or gastroesophageal junction (GEJ) cancer. Irinotecan 113-123 mitochondrially encoded cytochrome c oxidase I Homo sapiens 125-128 27654129-3 2017 Dose escalation of irinotecan was performed according to the results of UGT1A1 genotyping in all patients. Irinotecan 19-29 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 72-78 27878250-7 2017 Thus, the potential benefit of the use of a combination of camptothecin-11 with other chemical drugs with inhibitory effects on IL-8 expression, should be paid more attention in treating colon cancer. Irinotecan 59-74 C-X-C motif chemokine ligand 8 Homo sapiens 128-132 27883280-0 2017 Phase I study of irinotecan for previously treated lung cancer patients with the UGT1A1*28 or *6 polymorphism: Results of the Lung Oncology Group in Kyushu (LOGIK1004A). Irinotecan 17-27 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 81-87 27883280-1 2017 BACKGROUND: Various polymorphisms have been detected in the UDP-glucuronosyltransferase 1A ( UGT1A ) gene, and UGT1A1 *28 and UGT1A1 *6 have important effects on the pharmacokinetics of irinotecan and the risk of severe toxicities during irinotecan therapy. Irinotecan 186-196 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 60-90 27883280-1 2017 BACKGROUND: Various polymorphisms have been detected in the UDP-glucuronosyltransferase 1A ( UGT1A ) gene, and UGT1A1 *28 and UGT1A1 *6 have important effects on the pharmacokinetics of irinotecan and the risk of severe toxicities during irinotecan therapy. Irinotecan 186-196 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-98 27883280-1 2017 BACKGROUND: Various polymorphisms have been detected in the UDP-glucuronosyltransferase 1A ( UGT1A ) gene, and UGT1A1 *28 and UGT1A1 *6 have important effects on the pharmacokinetics of irinotecan and the risk of severe toxicities during irinotecan therapy. Irinotecan 186-196 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 111-117 27883280-1 2017 BACKGROUND: Various polymorphisms have been detected in the UDP-glucuronosyltransferase 1A ( UGT1A ) gene, and UGT1A1 *28 and UGT1A1 *6 have important effects on the pharmacokinetics of irinotecan and the risk of severe toxicities during irinotecan therapy. Irinotecan 186-196 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 126-132 27883280-1 2017 BACKGROUND: Various polymorphisms have been detected in the UDP-glucuronosyltransferase 1A ( UGT1A ) gene, and UGT1A1 *28 and UGT1A1 *6 have important effects on the pharmacokinetics of irinotecan and the risk of severe toxicities during irinotecan therapy. Irinotecan 238-248 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 60-90 27883280-1 2017 BACKGROUND: Various polymorphisms have been detected in the UDP-glucuronosyltransferase 1A ( UGT1A ) gene, and UGT1A1 *28 and UGT1A1 *6 have important effects on the pharmacokinetics of irinotecan and the risk of severe toxicities during irinotecan therapy. Irinotecan 238-248 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-98 27883280-1 2017 BACKGROUND: Various polymorphisms have been detected in the UDP-glucuronosyltransferase 1A ( UGT1A ) gene, and UGT1A1 *28 and UGT1A1 *6 have important effects on the pharmacokinetics of irinotecan and the risk of severe toxicities during irinotecan therapy. Irinotecan 238-248 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 111-117 27883280-1 2017 BACKGROUND: Various polymorphisms have been detected in the UDP-glucuronosyltransferase 1A ( UGT1A ) gene, and UGT1A1 *28 and UGT1A1 *6 have important effects on the pharmacokinetics of irinotecan and the risk of severe toxicities during irinotecan therapy. Irinotecan 238-248 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 126-132 27883280-2 2017 This study was conducted to determine the maximum tolerated dose (MTD) of irinotecan chemotherapy according to the UGT1A1 genotype in previously treated lung cancer patients with the UGT1A1 *28 or UGT1A1 *6 polymorphism. Irinotecan 74-84 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 115-121 27883280-2 2017 This study was conducted to determine the maximum tolerated dose (MTD) of irinotecan chemotherapy according to the UGT1A1 genotype in previously treated lung cancer patients with the UGT1A1 *28 or UGT1A1 *6 polymorphism. Irinotecan 74-84 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 183-189 27883280-2 2017 This study was conducted to determine the maximum tolerated dose (MTD) of irinotecan chemotherapy according to the UGT1A1 genotype in previously treated lung cancer patients with the UGT1A1 *28 or UGT1A1 *6 polymorphism. Irinotecan 74-84 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 183-189 27883280-10 2017 CONCLUSIONS: The MTD of irinotecan for previously treated lung cancer patients that are heterozygous for the UGT1A1 * 28 or UGT1A1 * 6 gene polymorphism is 60 mg/m2 . Irinotecan 24-34 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 109-115 27883280-10 2017 CONCLUSIONS: The MTD of irinotecan for previously treated lung cancer patients that are heterozygous for the UGT1A1 * 28 or UGT1A1 * 6 gene polymorphism is 60 mg/m2 . Irinotecan 24-34 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 124-130 27638859-0 2016 ATM Expression Predicts Veliparib and Irinotecan Sensitivity in Gastric Cancer by Mediating P53-Independent Regulation of Cell Cycle and Apoptosis. Irinotecan 38-48 ATM serine/threonine kinase Homo sapiens 0-3 27838229-0 2016 Carvacrol reduces irinotecan-induced intestinal mucositis through inhibition of inflammation and oxidative damage via TRPA1 receptor activation. Irinotecan 18-28 transient receptor potential cation channel, subfamily A, member 1 Mus musculus 118-123 27838229-4 2016 Thus, the aim of the present study was to verify the supposed anti-inflammatory and protective action of carvacrol via TRPA1 activation against intestinal mucositis induced by CPT-11 in mice. Irinotecan 176-182 transient receptor potential cation channel, subfamily A, member 1 Mus musculus 119-124 27838229-10 2016 Carvacrol was found to exert an anti-inflammatory action against CPT-11-induced intestinal mucositis through strong interactions with TRPA1 receptors; reduction in the production or release or both of pro-inflammatory cytokines (TNF-alpha, IL-1beta, and KC); and decrease in other indicators of inflammation (MPO, NF-kappaB, COX-2) and oxidative stress (GSH, MDA, and NOx levels). Irinotecan 65-71 transient receptor potential cation channel, subfamily A, member 1 Mus musculus 134-139 27838229-10 2016 Carvacrol was found to exert an anti-inflammatory action against CPT-11-induced intestinal mucositis through strong interactions with TRPA1 receptors; reduction in the production or release or both of pro-inflammatory cytokines (TNF-alpha, IL-1beta, and KC); and decrease in other indicators of inflammation (MPO, NF-kappaB, COX-2) and oxidative stress (GSH, MDA, and NOx levels). Irinotecan 65-71 interleukin 1 beta Mus musculus 240-248 27838229-10 2016 Carvacrol was found to exert an anti-inflammatory action against CPT-11-induced intestinal mucositis through strong interactions with TRPA1 receptors; reduction in the production or release or both of pro-inflammatory cytokines (TNF-alpha, IL-1beta, and KC); and decrease in other indicators of inflammation (MPO, NF-kappaB, COX-2) and oxidative stress (GSH, MDA, and NOx levels). Irinotecan 65-71 myeloperoxidase Mus musculus 309-312 27838229-10 2016 Carvacrol was found to exert an anti-inflammatory action against CPT-11-induced intestinal mucositis through strong interactions with TRPA1 receptors; reduction in the production or release or both of pro-inflammatory cytokines (TNF-alpha, IL-1beta, and KC); and decrease in other indicators of inflammation (MPO, NF-kappaB, COX-2) and oxidative stress (GSH, MDA, and NOx levels). Irinotecan 65-71 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 314-323 27838229-10 2016 Carvacrol was found to exert an anti-inflammatory action against CPT-11-induced intestinal mucositis through strong interactions with TRPA1 receptors; reduction in the production or release or both of pro-inflammatory cytokines (TNF-alpha, IL-1beta, and KC); and decrease in other indicators of inflammation (MPO, NF-kappaB, COX-2) and oxidative stress (GSH, MDA, and NOx levels). Irinotecan 65-71 cytochrome c oxidase II, mitochondrial Mus musculus 325-330 28118131-8 2016 The proposed method was successfully applied to the determination of irinotecan in tap water, river water, and urine samples spiked with 10.20 mg/L for the water samples and 8.32 mg/L for the urine sample. Irinotecan 69-79 nuclear RNA export factor 1 Homo sapiens 83-86 28118131-9 2016 The average recovery values of irinotecan determined were 99.1% for tap water, 109.4% for river water, and 96.1% for urine. Irinotecan 31-41 nuclear RNA export factor 1 Homo sapiens 68-71 27587582-7 2016 This is demonstrated with the discovery that HMGA2 potentiates the clinically important topoisomerase I inhibitor irinotecan/SN-38 in trapping the enzyme in covalent DNA-complexes, thereby attenuating transcription. Irinotecan 114-124 high mobility group AT-hook 2 Homo sapiens 45-50 27587582-7 2016 This is demonstrated with the discovery that HMGA2 potentiates the clinically important topoisomerase I inhibitor irinotecan/SN-38 in trapping the enzyme in covalent DNA-complexes, thereby attenuating transcription. Irinotecan 125-130 high mobility group AT-hook 2 Homo sapiens 45-50 28345025-4 2017 Human-derived NB cell lines were significantly more sensitive to treatment with hCE1m6-NSCs and irinotecan as compared with drug alone. Irinotecan 96-106 carboxylesterase 1 Homo sapiens 80-84 27729313-1 2016 In vitro, EGFR inhibition, combined with the BRAF inhibitor vemurafenib, causes synergistic cytotoxicity for BRAFV600E metastatic colorectal cancer, further augmented by irinotecan. Irinotecan 170-180 epidermal growth factor receptor Homo sapiens 10-14 27888624-0 2016 RhoA regulates resistance to irinotecan by regulating membrane transporter and apoptosis signaling in colorectal cancer. Irinotecan 29-39 ras homolog family member A Homo sapiens 0-4 27888624-4 2016 In this study, we show that, compared with parental cells, RhoA is up-regulated in irinotecan (CPT-11)-resistant CRC cells. Irinotecan 83-93 ras homolog family member A Homo sapiens 59-63 27888624-4 2016 In this study, we show that, compared with parental cells, RhoA is up-regulated in irinotecan (CPT-11)-resistant CRC cells. Irinotecan 95-101 ras homolog family member A Homo sapiens 59-63 27888624-5 2016 Furthermore, inhibition of RhoA in drug resistant cells, at least partially, rescues the resistance against irinotecan and increases the sensitivity to other chemotherapeutic drug by inhibiting expression of MDR1, MRP1and GSTP1, promotes apoptosis by suppressing the expression of BCL-XL and Bcl-2 and increasing Bax expression, and significantly decreases side population cells. Irinotecan 108-118 ras homolog family member A Homo sapiens 27-31 27960424-2 2016 TP1, consisting of the antineoplastic camptothecin analogue SN-38, and the fluorescent dye rhodol green have been covalently conjugated through a disulfide bond. Irinotecan 60-65 transition protein 1 Homo sapiens 0-3 26687137-0 2016 Complete Clinical Response of BRAF-Mutated Cholangiocarcinoma to Vemurafenib, Panitumumab, and Irinotecan. Irinotecan 95-105 B-Raf proto-oncogene, serine/threonine kinase Homo sapiens 30-34 27638859-4 2016 The present study evaluated ATM-induced synthetic lethality and its role in sensitization of gastric cancer cells to PARP and TOP1 inhibitors, veliparib (ABT-888) and irinotecan (CPT-11), respectively. Irinotecan 167-177 ATM serine/threonine kinase Homo sapiens 28-31 27638859-4 2016 The present study evaluated ATM-induced synthetic lethality and its role in sensitization of gastric cancer cells to PARP and TOP1 inhibitors, veliparib (ABT-888) and irinotecan (CPT-11), respectively. Irinotecan 179-185 ATM serine/threonine kinase Homo sapiens 28-31 27638859-6 2016 The combinatorial effect of ABT-888 and CPT-11 in gastric cancer cells was also determined both in vitro and in vivo ATM deficiency was found to be associated with enhanced sensitivity to ABT-888 and CPT-11 monotherapy, hence suggesting a mechanism of synthetic lethality. Irinotecan 40-46 ATM serine/threonine kinase Homo sapiens 117-120 27638859-6 2016 The combinatorial effect of ABT-888 and CPT-11 in gastric cancer cells was also determined both in vitro and in vivo ATM deficiency was found to be associated with enhanced sensitivity to ABT-888 and CPT-11 monotherapy, hence suggesting a mechanism of synthetic lethality. Irinotecan 200-206 ATM serine/threonine kinase Homo sapiens 117-120 27638859-7 2016 Cells with high ATM expression showed reduced sensitivity to monotherapy; however, they showed a higher therapeutic effect with ABT-888 and CPT-11 combinatorial therapy. Irinotecan 140-146 ATM serine/threonine kinase Homo sapiens 16-19 27550942-0 2016 TLR4-Dependent Claudin-1 Internalization and Secretagogue-Mediated Chloride Secretion Regulate Irinotecan-Induced Diarrhea. Irinotecan 95-105 toll-like receptor 4 Mus musculus 0-4 27697767-9 2016 The net result of combined lurbinectedin and irinotecan treatment was a complete reversal of EWS-FLI1 activity and elimination of established tumors in 30% to 70% of mice after only 11 days of therapy. Irinotecan 45-55 Ewing sarcoma breakpoint region 1 Mus musculus 93-96 27697767-9 2016 The net result of combined lurbinectedin and irinotecan treatment was a complete reversal of EWS-FLI1 activity and elimination of established tumors in 30% to 70% of mice after only 11 days of therapy. Irinotecan 45-55 Friend leukemia integration 1 Mus musculus 97-101 27546072-8 2016 This review aims to summarize and evaluate the implications of pharmacogenetic variants in the ABC and SLC transporters genes on the pharmacokinetics and clinical outcomes of three anti-cancer agents: irinotecan, docetaxel and doxorubicin in Caucasian and Asian patients. Irinotecan 201-211 C-C motif chemokine ligand 21 Homo sapiens 103-106 28133330-5 2016 S-1/irinotecan combination chemotherapy was administered as second-line chemotherapy, but he developed grade 3 diarrhea, and the S-1/irinotecan combination chemotherapy was immediately stopped.Weekly paclitaxel chemother- apy was administered as third-line chemotherapy, and S-1/docetaxel combination chemotherapy was administered as fourth-line chemotherapy. Irinotecan 133-143 proteasome 26S subunit, non-ATPase 1 Homo sapiens 0-3 27550942-0 2016 TLR4-Dependent Claudin-1 Internalization and Secretagogue-Mediated Chloride Secretion Regulate Irinotecan-Induced Diarrhea. Irinotecan 95-105 claudin 1 Mus musculus 15-24 27550942-8 2016 Irinotecan caused internalization of claudin-1 with focal lesions of ZO-1 and occludin proteolysis in the ileum and colon of wild-type mice. Irinotecan 0-10 claudin 1 Mus musculus 37-46 27550942-8 2016 Irinotecan caused internalization of claudin-1 with focal lesions of ZO-1 and occludin proteolysis in the ileum and colon of wild-type mice. Irinotecan 0-10 tight junction protein 1 Mus musculus 69-73 27550942-12 2016 Increased carbachol-induced chloride secretion was seen in irinotecan-treated wild-type and Tlr4-/- mice at 24 hours (wild-type: 100.35 +- 18.37 muA/cm2; P = 0.022; Tlr4-/-: 102.72 +- 18.80 muA/cm2; P = 0.023). Irinotecan 59-69 toll-like receptor 4 Mus musculus 92-96 27550942-12 2016 Increased carbachol-induced chloride secretion was seen in irinotecan-treated wild-type and Tlr4-/- mice at 24 hours (wild-type: 100.35 +- 18.37 muA/cm2; P = 0.022; Tlr4-/-: 102.72 +- 18.80 muA/cm2; P = 0.023). Irinotecan 59-69 toll-like receptor 4 Mus musculus 165-169 27748862-0 2016 Gap junction composed of connexin43 modulates 5-fluorouracil, oxaliplatin and irinotecan resistance on colorectal cancers. Irinotecan 78-88 gap junction protein alpha 1 Homo sapiens 25-35 27550942-13 2016 Results suggest that TLR4-dependent claudin-1 internalization and secondary anion secretion contribute to irinotecan-induced diarrhea. Irinotecan 106-116 toll-like receptor 4 Mus musculus 21-25 27748862-4 2016 The current study investigated the effects of connexin43 (Cx43) gap junctions on 5-fluorouracil (5-FU), oxaliplatin and irinotecan in colon cancer cells. Irinotecan 120-130 gap junction protein alpha 1 Homo sapiens 58-62 27550942-13 2016 Results suggest that TLR4-dependent claudin-1 internalization and secondary anion secretion contribute to irinotecan-induced diarrhea. Irinotecan 106-116 claudin 1 Mus musculus 36-45 27748862-7 2016 Downregulation of Cx43 gap junction functioning attenuated 5-FU, oxaliplatin and irinotecan toxicity in colon cancer cells, which was increased in cells treated with a Cx43 gap junction function enhancer. Irinotecan 81-91 gap junction protein alpha 1 Homo sapiens 18-22 27748862-7 2016 Downregulation of Cx43 gap junction functioning attenuated 5-FU, oxaliplatin and irinotecan toxicity in colon cancer cells, which was increased in cells treated with a Cx43 gap junction function enhancer. Irinotecan 81-91 gap junction protein alpha 1 Homo sapiens 168-172 30550693-3 2016 Recent pharmacogenetic studies on irinotecan have revealed the impact of UGT1A1 polymorphisms on severe adverse effects. Irinotecan 34-44 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 73-79 27895797-4 2016 The promoter (-1000 bp) and exon 1 regions of UDP glucuronosyltransferase family 1 member A complex locus (UGT1A1) gene family members UGT1A1, UGT1A7 and UGT1A9 were sequenced, and comprehensive analysis of their genetic polymorphisms was performed to determine the association between inherited genetic variations and irinotecan-induced toxicity. Irinotecan 319-329 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 135-141 27895797-6 2016 The results of the present study revealed that UGT1A1*6 and UGT1A7*3 are risk factors for irinotecan-induced severe neutropenia, and UGT1A9*1b is associated with severe diarrhea. Irinotecan 90-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 47-53 27895797-6 2016 The results of the present study revealed that UGT1A1*6 and UGT1A7*3 are risk factors for irinotecan-induced severe neutropenia, and UGT1A9*1b is associated with severe diarrhea. Irinotecan 90-100 UDP glucuronosyltransferase family 1 member A7 Homo sapiens 60-66 27895797-0 2016 UGT1A1*6, UGT1A7*3 and UGT1A9*1b polymorphisms are predictive markers for severe toxicity in patients with metastatic gastrointestinal cancer treated with irinotecan-based regimens. Irinotecan 155-165 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 27895797-0 2016 UGT1A1*6, UGT1A7*3 and UGT1A9*1b polymorphisms are predictive markers for severe toxicity in patients with metastatic gastrointestinal cancer treated with irinotecan-based regimens. Irinotecan 155-165 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 23-29 27895797-4 2016 The promoter (-1000 bp) and exon 1 regions of UDP glucuronosyltransferase family 1 member A complex locus (UGT1A1) gene family members UGT1A1, UGT1A7 and UGT1A9 were sequenced, and comprehensive analysis of their genetic polymorphisms was performed to determine the association between inherited genetic variations and irinotecan-induced toxicity. Irinotecan 319-329 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 107-113 30550693-4 2016 The concurrence of UGT1A1*28 and UGT1A1*6, even when heterozygous, markedly alters the disposition of irinotecan, potentially increasing toxicity. Irinotecan 102-112 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 19-25 30550693-4 2016 The concurrence of UGT1A1*28 and UGT1A1*6, even when heterozygous, markedly alters the disposition of irinotecan, potentially increasing toxicity. Irinotecan 102-112 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-39 30550693-5 2016 For patients showing homozygosity for UGT1A1*28, *6 or compound heterozygosity for UGT1A1*6 and *28, dose reduction of irinotecan is strongly recommended. Irinotecan 119-129 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 38-44 30550693-5 2016 For patients showing homozygosity for UGT1A1*28, *6 or compound heterozygosity for UGT1A1*6 and *28, dose reduction of irinotecan is strongly recommended. Irinotecan 119-129 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 83-89 27770825-0 2016 Suppression of lupus nephritis and skin lesions in MRL/lpr mice by administration of the topoisomerase I inhibitor irinotecan. Irinotecan 115-125 Fas (TNF receptor superfamily member 6) Mus musculus 55-58 27770825-8 2016 RESULTS: Administration of both high- and low-dose irinotecan prevented proteinuria and prolonged survival in MRL/lpr mice. Irinotecan 51-61 Fas (TNF receptor superfamily member 6) Mus musculus 114-117 27115099-9 2016 Among these, the irinotecan/temozolomide combination induced strong tumor regression in the TCLT and in a model derived from an AFP-negative relapsing HB. Irinotecan 17-27 alpha fetoprotein Mus musculus 128-131 27267850-5 2016 EXPERIMENTAL DESIGN: To avoid DLT for useful cytotoxic agents, the recently developed drug STA-8666 combines a chemical moiety targeting active HSP90 (concentrated in tumors) fused via cleavable linker to SN38, the active metabolite of irinotecan. Irinotecan 236-246 GCY Homo sapiens 91-94 27267850-6 2016 We compare potency and mechanism of action of STA-8666 and irinotecan in vitro and in vivo RESULTS: In two SCLC xenograft and patient-derived xenograft models, STA-8666 was tolerated without side effects up to 150 mg/kg. Irinotecan 59-69 GCY Homo sapiens 160-163 27510985-0 2016 Effect of hesperidin on the pharmacokinetics of CPT-11 and its active metabolite SN-38 by regulating hepatic Mrp2 in rats. Irinotecan 48-54 ATP binding cassette subfamily C member 2 Rattus norvegicus 109-113 27510985-0 2016 Effect of hesperidin on the pharmacokinetics of CPT-11 and its active metabolite SN-38 by regulating hepatic Mrp2 in rats. Irinotecan 81-86 ATP binding cassette subfamily C member 2 Rattus norvegicus 109-113 27510985-3 2016 Accumulated evidence showed that the regulatory effect of hesperidin on the expression of Mrp2 in the liver may be one of the critical factors controlling the biliary excretion of SN-38. Irinotecan 180-185 ATP binding cassette subfamily C member 2 Rattus norvegicus 90-94 27399284-4 2016 FANCJ interacts with HP1gamma in a BARD1-dependent manner, and this interaction was enhanced by ionizing radiation or irinotecan hydrochloride treatment. Irinotecan 118-142 BRCA1 interacting helicase 1 Homo sapiens 0-5 27399284-4 2016 FANCJ interacts with HP1gamma in a BARD1-dependent manner, and this interaction was enhanced by ionizing radiation or irinotecan hydrochloride treatment. Irinotecan 118-142 chromobox 3 Homo sapiens 21-29 27399284-4 2016 FANCJ interacts with HP1gamma in a BARD1-dependent manner, and this interaction was enhanced by ionizing radiation or irinotecan hydrochloride treatment. Irinotecan 118-142 BRCA1 associated RING domain 1 Homo sapiens 35-40 27716027-6 2016 Both Irinotecan/HAS2 (Hyaluronan synthase 2) and Bevacizumab/PGAM1 (Phosphoglycerate mutase 1) are interactions found in this study with independent confirmation. Irinotecan 5-15 hyaluronan synthase 2 Homo sapiens 16-20 27716027-6 2016 Both Irinotecan/HAS2 (Hyaluronan synthase 2) and Bevacizumab/PGAM1 (Phosphoglycerate mutase 1) are interactions found in this study with independent confirmation. Irinotecan 5-15 hyaluronan synthase 2 Homo sapiens 22-43 27716027-6 2016 Both Irinotecan/HAS2 (Hyaluronan synthase 2) and Bevacizumab/PGAM1 (Phosphoglycerate mutase 1) are interactions found in this study with independent confirmation. Irinotecan 5-15 phosphoglycerate mutase 1 Homo sapiens 61-66 27716027-6 2016 Both Irinotecan/HAS2 (Hyaluronan synthase 2) and Bevacizumab/PGAM1 (Phosphoglycerate mutase 1) are interactions found in this study with independent confirmation. Irinotecan 5-15 phosphoglycerate mutase 1 Homo sapiens 68-93 27608846-0 2016 STA-8666, a novel HSP90 inhibitor/SN-38 drug conjugate, causes complete tumor regression in preclinical mouse models of pediatric sarcoma. Irinotecan 34-39 autosomal striping Mus musculus 0-3 27608846-3 2016 In this study we have evaluated the efficacy and toxicity of STA-8666, a novel drug conjugate which uses an HSP90 inhibitor to facilitate intracellular, tumor-targeted delivery of the topoisomerase 1 inhibitor SN-38, thus preferentially delivering and concentrating SN-38 within tumor tissue. Irinotecan 210-215 autosomal striping Mus musculus 61-64 27608846-3 2016 In this study we have evaluated the efficacy and toxicity of STA-8666, a novel drug conjugate which uses an HSP90 inhibitor to facilitate intracellular, tumor-targeted delivery of the topoisomerase 1 inhibitor SN-38, thus preferentially delivering and concentrating SN-38 within tumor tissue. Irinotecan 210-215 heat shock protein 86, pseudogene 1 Mus musculus 108-113 27608846-3 2016 In this study we have evaluated the efficacy and toxicity of STA-8666, a novel drug conjugate which uses an HSP90 inhibitor to facilitate intracellular, tumor-targeted delivery of the topoisomerase 1 inhibitor SN-38, thus preferentially delivering and concentrating SN-38 within tumor tissue. Irinotecan 266-271 autosomal striping Mus musculus 61-64 27608846-6 2016 Gene expression analysis performed on xenograft tumors treated with either irinotecan or STA-8666 showed that STA-8666 affected expression of DNA damage and repair genes more robustly than irinotecan. Irinotecan 75-85 autosomal striping Mus musculus 110-113 27608846-6 2016 Gene expression analysis performed on xenograft tumors treated with either irinotecan or STA-8666 showed that STA-8666 affected expression of DNA damage and repair genes more robustly than irinotecan. Irinotecan 189-199 autosomal striping Mus musculus 89-92 27116457-0 2016 Effect of Single Nucleotide Polymorphisms in the Xenobiotic-sensing Receptors NR1I2 and NR1I3 on the Pharmacokinetics and Toxicity of Irinotecan in Colorectal Cancer Patients. Irinotecan 134-144 nuclear receptor subfamily 1 group I member 2 Homo sapiens 78-83 27698852-0 2016 ABCG2 expression in colorectal adenocarcinomas may predict resistance to irinotecan. Irinotecan 73-83 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 0-5 27698852-3 2016 The aim of the present study was to evaluate whether the expression levels of adenosine triphosphate-binding cassette sub-family G (WHITE) member 2 (Junior blood group) (ABCG2) in primary colorectal tumors predict chemoresistance to irinotecan. Irinotecan 233-243 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 170-175 27698852-4 2016 Using the resected primary tumor specimens of 189 patients with colorectal cancer, the association between the immunohistochemical expression of ABCG2 protein and the results of the collagen gel droplet embedded culture drug sensitivity test, performed to evaluate the chemosensitivity to SN-38 (an active metabolite of irinotecan), was investigated. Irinotecan 289-294 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 145-150 27698852-4 2016 Using the resected primary tumor specimens of 189 patients with colorectal cancer, the association between the immunohistochemical expression of ABCG2 protein and the results of the collagen gel droplet embedded culture drug sensitivity test, performed to evaluate the chemosensitivity to SN-38 (an active metabolite of irinotecan), was investigated. Irinotecan 320-330 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 145-150 27698852-6 2016 The tumors of patients with increased ABCG2 expression accounted for 60% of the tumors examined, and were significantly more resistant to SN-38, compared with patients with low ABCG2 expression (P<0.001). Irinotecan 138-143 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 38-43 27698852-7 2016 In a multivariate logistic regression analysis, increased expression of ABCG2 protein was an independent and significant predictor of resistance to SN-38, increasing the risk of resistance by 12-fold. Irinotecan 148-153 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 72-77 27698852-8 2016 Increased expression of ABCG2 and a low sensitivity to SN-38 was significantly associated with resistance to irinotecan-based chemotherapy (P=0.01 and 0.028, respectively). Irinotecan 109-119 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 24-29 27698852-10 2016 The increased immunohistochemical expression of ABCG2 in primary tumors may be a useful predictive biomarker of resistance to irinotecan-based chemotherapy for patients with recurrent or metastatic colorectal cancer. Irinotecan 126-136 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 48-53 27121793-1 2016 PURPOSE: WRN promoter CpG island hypermethylation in colorectal cancer has been reported to increase sensitivity to irinotecan-based therapies. Irinotecan 116-126 WRN RecQ like helicase Homo sapiens 9-12 27468920-1 2016 PURPOSE: Anti-epidermal growth factor receptor antibody therapy alone or in combination with irinotecan is recognized as a standard third-line treatment for KRAS wild-type unresectable metastatic colorectal cancer. Irinotecan 93-103 KRAS proto-oncogene, GTPase Homo sapiens 157-161 27468920-13 2016 CONCLUSIONS: Combination therapy with cetuximab and S-1 was effective and well tolerated in patients with irinotecan-, oxaliplatin-, and fluoropyrimidine-refractory metastatic colorectal cancer. Irinotecan 106-116 proteasome 26S subunit, non-ATPase 1 Homo sapiens 52-55 27116457-0 2016 Effect of Single Nucleotide Polymorphisms in the Xenobiotic-sensing Receptors NR1I2 and NR1I3 on the Pharmacokinetics and Toxicity of Irinotecan in Colorectal Cancer Patients. Irinotecan 134-144 nuclear receptor subfamily 1 group I member 3 Homo sapiens 88-93 27116457-1 2016 BACKGROUND AND OBJECTIVES: Nuclear receptors PXR (pregnane X receptor, NR1I2) and CAR (constitutive androstane receptor, NR1I3) are key regulators of irinotecan metabolism, and ligand-dependent modulation of their activity leads to significant drug-drug interactions. Irinotecan 150-160 nuclear receptor subfamily 1 group I member 2 Homo sapiens 45-48 27116457-1 2016 BACKGROUND AND OBJECTIVES: Nuclear receptors PXR (pregnane X receptor, NR1I2) and CAR (constitutive androstane receptor, NR1I3) are key regulators of irinotecan metabolism, and ligand-dependent modulation of their activity leads to significant drug-drug interactions. Irinotecan 150-160 nuclear receptor subfamily 1 group I member 2 Homo sapiens 50-69 27116457-1 2016 BACKGROUND AND OBJECTIVES: Nuclear receptors PXR (pregnane X receptor, NR1I2) and CAR (constitutive androstane receptor, NR1I3) are key regulators of irinotecan metabolism, and ligand-dependent modulation of their activity leads to significant drug-drug interactions. Irinotecan 150-160 nuclear receptor subfamily 1 group I member 2 Homo sapiens 71-76 27116457-1 2016 BACKGROUND AND OBJECTIVES: Nuclear receptors PXR (pregnane X receptor, NR1I2) and CAR (constitutive androstane receptor, NR1I3) are key regulators of irinotecan metabolism, and ligand-dependent modulation of their activity leads to significant drug-drug interactions. Irinotecan 150-160 nuclear receptor subfamily 1 group I member 3 Homo sapiens 82-126 27116457-4 2016 RESULTS: After adjustment of the tests by the UGT1A1*28 genotype and correction for multiple testing, the A allele of NR1I2-rs10934498 was associated with a decreased exposition and an increased degradation of SN-38, the active metabolite (p = 0.009 and p = 0.017, respectively). Irinotecan 210-215 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 46-52 27116457-4 2016 RESULTS: After adjustment of the tests by the UGT1A1*28 genotype and correction for multiple testing, the A allele of NR1I2-rs10934498 was associated with a decreased exposition and an increased degradation of SN-38, the active metabolite (p = 0.009 and p = 0.017, respectively). Irinotecan 210-215 nuclear receptor subfamily 1 group I member 2 Homo sapiens 118-123 27116457-6 2016 CONCLUSION: Our results reveal for the first time the involvement of NR1I2 in the pharmacogenetics of irinotecan and suggest that it may help to predict the toxicity of low-dose irinotecan. Irinotecan 102-112 nuclear receptor subfamily 1 group I member 2 Homo sapiens 69-74 27116457-6 2016 CONCLUSION: Our results reveal for the first time the involvement of NR1I2 in the pharmacogenetics of irinotecan and suggest that it may help to predict the toxicity of low-dose irinotecan. Irinotecan 178-188 nuclear receptor subfamily 1 group I member 2 Homo sapiens 69-74 26750665-7 2016 Moreover, the GA-mediated repression of CES2 attenuated CPT-11-induced apoptosis. Irinotecan 56-62 carboxylesterase 2 Homo sapiens 40-44 27338835-7 2016 NDRG1 was a favorable prognostic factor of mCRC, although might be responsible for CPT-11 resistance in vitro. Irinotecan 83-89 N-myc downstream regulated 1 Homo sapiens 0-5 26744836-12 2016 The substrate uptakes of OAT1, OCTN1 and OCTN2 were significantly decreased in the presence of CZ112, while CPT-11 potently down-regulated the transport activity of OCT1 and OCT3. Irinotecan 108-114 solute carrier family 22 member 1 Homo sapiens 165-169 27207776-0 2016 Combining ABCG2 Inhibitors with IMMU-132, an Anti-Trop-2 Antibody Conjugate of SN-38, Overcomes Resistance to SN-38 in Breast and Gastric Cancers. Irinotecan 79-84 tumor associated calcium signal transducer 2 Homo sapiens 50-56 27380166-0 2016 Erratum to: UDP-glucuronosyltransferase 1A1*6 and *28 polymorphisms as indicators of initial dose level of irinotecan to reduce risk of neutropenia in patients receiving FOLFIRI for colorectal cancer. Irinotecan 107-117 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-43 26950035-0 2016 MRP1 expression in CTCs confers resistance to irinotecan-based chemotherapy in metastatic colorectal cancer. Irinotecan 46-56 ATP binding cassette subfamily C member 1 Homo sapiens 0-4 26950035-3 2016 Multidrug resistance-associated protein 1 (MRP1) and Multidrug resistance-associated protein 4 (MRP4) play a role in irinotecan-resistance, and Excision Repair Cross-Complementation group 1 (ERCC1) expression can confer resistance to platinum compounds. Irinotecan 117-127 ATP binding cassette subfamily C member 1 Homo sapiens 0-41 26950035-3 2016 Multidrug resistance-associated protein 1 (MRP1) and Multidrug resistance-associated protein 4 (MRP4) play a role in irinotecan-resistance, and Excision Repair Cross-Complementation group 1 (ERCC1) expression can confer resistance to platinum compounds. Irinotecan 117-127 ATP binding cassette subfamily C member 1 Homo sapiens 43-47 26950035-3 2016 Multidrug resistance-associated protein 1 (MRP1) and Multidrug resistance-associated protein 4 (MRP4) play a role in irinotecan-resistance, and Excision Repair Cross-Complementation group 1 (ERCC1) expression can confer resistance to platinum compounds. Irinotecan 117-127 ATP binding cassette subfamily C member 4 Homo sapiens 53-94 26950035-3 2016 Multidrug resistance-associated protein 1 (MRP1) and Multidrug resistance-associated protein 4 (MRP4) play a role in irinotecan-resistance, and Excision Repair Cross-Complementation group 1 (ERCC1) expression can confer resistance to platinum compounds. Irinotecan 117-127 ATP binding cassette subfamily C member 4 Homo sapiens 96-100 26950035-3 2016 Multidrug resistance-associated protein 1 (MRP1) and Multidrug resistance-associated protein 4 (MRP4) play a role in irinotecan-resistance, and Excision Repair Cross-Complementation group 1 (ERCC1) expression can confer resistance to platinum compounds. Irinotecan 117-127 ERCC excision repair 1, endonuclease non-catalytic subunit Homo sapiens 191-196 26950035-7 2016 Among patients treated with irinotecan-based chemotherapy, 4 out of 19 cases with MRP1 positive CTCs showed a worse progression free survival (PFS) in comparison to those with MRP1 negative CTCs (2.1 months vs. 9.1 months; p = 0.003). Irinotecan 28-38 ATP binding cassette subfamily C member 1 Homo sapiens 82-86 26950035-7 2016 Among patients treated with irinotecan-based chemotherapy, 4 out of 19 cases with MRP1 positive CTCs showed a worse progression free survival (PFS) in comparison to those with MRP1 negative CTCs (2.1 months vs. 9.1 months; p = 0.003). Irinotecan 28-38 ATP binding cassette subfamily C member 1 Homo sapiens 176-180 26950035-10 2016 Our results show MRP1 as a potential biomarker of resistance to treatment with irinotecan when found in CTCs from mCRC patients. Irinotecan 79-89 ATP binding cassette subfamily C member 1 Homo sapiens 17-21 26710796-0 2016 UDP-glucuronosyltransferase 1A1*6 and *28 polymorphisms as indicators of initial dose level of irinotecan to reduce risk of neutropenia in patients receiving FOLFIRI for colorectal cancer. Irinotecan 95-105 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-31 27207776-0 2016 Combining ABCG2 Inhibitors with IMMU-132, an Anti-Trop-2 Antibody Conjugate of SN-38, Overcomes Resistance to SN-38 in Breast and Gastric Cancers. Irinotecan 110-115 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 10-15 27207776-0 2016 Combining ABCG2 Inhibitors with IMMU-132, an Anti-Trop-2 Antibody Conjugate of SN-38, Overcomes Resistance to SN-38 in Breast and Gastric Cancers. Irinotecan 110-115 tumor associated calcium signal transducer 2 Homo sapiens 50-56 27207776-6 2016 Importantly, treatment of both S120 sublines with known ABCG2 inhibitors (fumitremorgin C, Ko143, and YHO-13351) restored toxicity of SN-38, and the combination of YHO-13351 with IMMU-132 increased the median survival of mice bearing NCI-N87-S120 xenografts. Irinotecan 134-139 ATP binding cassette subfamily G member 2 (Junior blood group) Mus musculus 56-61 27006309-10 2016 The median progression-free survival was significantly different between groups with low or high MGMT expression for the irinotecan-based regimen (p = 0.025), but MGMT protein expression was not observed to be a prognostic factor. Irinotecan 121-131 O-6-methylguanine-DNA methyltransferase Homo sapiens 97-101 26739304-0 2016 High Resectability Rate of Initially Unresectable Colorectal Liver Metastases After UGT1A1-Adapted High-Dose Irinotecan Combined with LV5FU2 and Cetuximab: A Multicenter Phase II Study (ERBIFORT). Irinotecan 109-119 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 84-90 27348241-0 2016 Targeting Colorectal Cancer Stem-Like Cells with Anti-CD133 Antibody-Conjugated SN-38 Nanoparticles. Irinotecan 80-85 prominin 1 Homo sapiens 54-59 27348241-5 2016 To resolve the problem of chemotherapy failure, SN-38-loaded nanoparticles were conjugated with anti-CD133 antibody to target CD133-positive (CD133(+)) cells. Irinotecan 48-53 prominin 1 Homo sapiens 101-106 27348241-5 2016 To resolve the problem of chemotherapy failure, SN-38-loaded nanoparticles were conjugated with anti-CD133 antibody to target CD133-positive (CD133(+)) cells. Irinotecan 48-53 prominin 1 Homo sapiens 126-131 27348241-5 2016 To resolve the problem of chemotherapy failure, SN-38-loaded nanoparticles were conjugated with anti-CD133 antibody to target CD133-positive (CD133(+)) cells. Irinotecan 48-53 prominin 1 Homo sapiens 126-131 27348241-6 2016 In this study, anti-CD133 antibody-conjugated SN-38-loaded nanoparticles (CD133Ab-NPs-SN-38) efficiently bound to HCT116 cells, which overexpress CD133 glycoprotein. Irinotecan 46-51 prominin 1 Homo sapiens 20-25 27348241-6 2016 In this study, anti-CD133 antibody-conjugated SN-38-loaded nanoparticles (CD133Ab-NPs-SN-38) efficiently bound to HCT116 cells, which overexpress CD133 glycoprotein. Irinotecan 46-51 prominin 1 Homo sapiens 74-79 27348241-6 2016 In this study, anti-CD133 antibody-conjugated SN-38-loaded nanoparticles (CD133Ab-NPs-SN-38) efficiently bound to HCT116 cells, which overexpress CD133 glycoprotein. Irinotecan 86-91 prominin 1 Homo sapiens 20-25 27348241-6 2016 In this study, anti-CD133 antibody-conjugated SN-38-loaded nanoparticles (CD133Ab-NPs-SN-38) efficiently bound to HCT116 cells, which overexpress CD133 glycoprotein. Irinotecan 86-91 prominin 1 Homo sapiens 74-79 27348241-8 2016 Furthermore, CD133Ab-NPs-SN-38 could target CD133(+) cells and inhibit colony formation compared with NPs-SN-38. Irinotecan 25-30 prominin 1 Homo sapiens 13-18 27348241-10 2016 A reduction in CD133 expression in HCT116 cells treated with CD133Ab-NPs-SN-38 was also observed in immunohistochemistry results. Irinotecan 73-78 prominin 1 Homo sapiens 15-20 27280693-8 2016 Furthermore, they are able to reverse the ABCG2-mediated resistance toward SN-38 and inhibit the ATPase activity. Irinotecan 75-80 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 42-47 27354601-0 2016 Early Experience with 70-150 mum Irinotecan Drug-eluting Beads (M1-DEBIRI) for the Treatment of Unresectable Hepatic Colorectal Metastases. Irinotecan 33-43 myoregulin Homo sapiens 64-73 27354601-1 2016 AIM: To report our early experience on the feasibility and safety of 70-150 mum drug-eluting beads loaded with irinotecan (M1-DEBIRI) for treating unresectable hepatic colorectal metastases. Irinotecan 111-121 myoregulin Homo sapiens 123-132 27286453-0 2016 UCN-01 enhances cytotoxicity of irinotecan in colorectal cancer stem-like cells by impairing DNA damage response. Irinotecan 32-42 urocortin Homo sapiens 0-3 27286453-5 2016 Nonetheless, broad-spectrum inhibition by the staurosporin derivative UCN-01 blocks CRC-SC growth and potentiates the activity of irinotecan in vitro and in vivo CRC-SC-derived models. Irinotecan 130-140 urocortin Homo sapiens 70-73 27286453-7 2016 Combination of LY2603618, a specific Chk1/2 inhibitor, with irinotecan resulted in a significant reduction of CRC-SC growth in vivo, confirming that irinotecan treatment coupled to inhibition of Chk1 represents a potentially effective therapeutic approach for CRC treatment. Irinotecan 149-159 checkpoint kinase 1 Homo sapiens 37-43 27286453-7 2016 Combination of LY2603618, a specific Chk1/2 inhibitor, with irinotecan resulted in a significant reduction of CRC-SC growth in vivo, confirming that irinotecan treatment coupled to inhibition of Chk1 represents a potentially effective therapeutic approach for CRC treatment. Irinotecan 149-159 checkpoint kinase 1 Homo sapiens 37-41 27354619-0 2016 Comparison of Panitumumab Plus Irinotecan and Cetuximab Plus Irinotecan for KRAS Wild-type Metastatic Colorectal Cancer. Irinotecan 61-71 KRAS proto-oncogene, GTPase Homo sapiens 76-80 26739304-14 2016 It seemed to be well-tolerated among healthy selected patients thanks to irinotecan dose adaptation according to UGT1A1 pharmacogenomics status. Irinotecan 73-83 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 113-119 27220761-0 2016 UGT1A1 gene polymorphism is associated with toxicity and clinical efficacy of irinotecan-based chemotherapy in patients with advanced colorectal cancer. Irinotecan 78-88 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 27220761-1 2016 OBJECTIVES: To investigate the relationship between uridine diphosphate glucoronosyltransferase 1A1 (UGT1A1)*28/*6 and toxicity and clinical efficacy of irinotecan-based chemotherapy in patients with advanced colorectal cancer (CRC) in Xinjiang Uygur and Han population. Irinotecan 153-163 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 52-99 27220761-1 2016 OBJECTIVES: To investigate the relationship between uridine diphosphate glucoronosyltransferase 1A1 (UGT1A1)*28/*6 and toxicity and clinical efficacy of irinotecan-based chemotherapy in patients with advanced colorectal cancer (CRC) in Xinjiang Uygur and Han population. Irinotecan 153-163 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 101-107 27220761-11 2016 CONCLUSION: UGT1A1 gene polymorphism predicts irinotecan-related adverse reactions in advanced CRC patients of Xinjiang Uygur and Han nationality; UGT1A1 gene polymorphism is correlated with efficacy and prognosis in Uygur nationality, but only related to prognosis in Han nationality in irinotecan-based chemotherapy. Irinotecan 46-56 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-18 27220761-11 2016 CONCLUSION: UGT1A1 gene polymorphism predicts irinotecan-related adverse reactions in advanced CRC patients of Xinjiang Uygur and Han nationality; UGT1A1 gene polymorphism is correlated with efficacy and prognosis in Uygur nationality, but only related to prognosis in Han nationality in irinotecan-based chemotherapy. Irinotecan 288-298 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-18 27220761-11 2016 CONCLUSION: UGT1A1 gene polymorphism predicts irinotecan-related adverse reactions in advanced CRC patients of Xinjiang Uygur and Han nationality; UGT1A1 gene polymorphism is correlated with efficacy and prognosis in Uygur nationality, but only related to prognosis in Han nationality in irinotecan-based chemotherapy. Irinotecan 288-298 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 147-153 27160286-0 2016 A novel genetic score model of UGT1A1 and TGFB pathway as predictor of severe irinotecan-related diarrhea in metastatic colorectal cancer patients. Irinotecan 78-88 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 31-37 26842236-15 2016 CONCLUSIONS: Veliparib can be safely combined with irinotecan at doses that inhibit PARP catalytic activity. Irinotecan 51-61 poly(ADP-ribose) polymerase 1 Homo sapiens 84-88 27101738-9 2016 D5D knockdown along with DGLA treatment also enhanced the cytotoxicities of various chemotherapeutic drugs, including 5-fluorouracil, regorafenib, and irinotecan, potentially through the activation of pro-apoptotic proteins, e.g. p53 and caspase 9. Irinotecan 151-161 fatty acid desaturase 1 Homo sapiens 0-3 26891420-1 2016 Cetuximab in combination with an irinotecan-containing regimen is a standard treatment in patients with KRAS wild-type (KRAS WT), metastatic colorectal cancer (mCRC). Irinotecan 33-43 KRAS proto-oncogene, GTPase Homo sapiens 104-108 27160286-0 2016 A novel genetic score model of UGT1A1 and TGFB pathway as predictor of severe irinotecan-related diarrhea in metastatic colorectal cancer patients. Irinotecan 78-88 transforming growth factor beta 1 Homo sapiens 42-46 26309154-0 2016 Biophysical and molecular docking insight into interaction mechanism and thermal stability of human serum albumin isoforms with a semi-synthetic water-soluble camptothecin analog irinotecan hydrochloride. Irinotecan 179-203 albumin Homo sapiens 100-113 26309154-1 2016 In the present work, we have examined the binding parameters, thermodynamics, and stability of human serum albumin (HSA) isoforms at pH 7.4 and 9.0, using spectroscopic, calorimetric, and molecular docking methods in the presence of water-soluble camptothecin analog irinotecan hydrochloride (CPT-11). Irinotecan 267-291 albumin Homo sapiens 101-114 26309154-1 2016 In the present work, we have examined the binding parameters, thermodynamics, and stability of human serum albumin (HSA) isoforms at pH 7.4 and 9.0, using spectroscopic, calorimetric, and molecular docking methods in the presence of water-soluble camptothecin analog irinotecan hydrochloride (CPT-11). Irinotecan 293-299 albumin Homo sapiens 101-114 27160286-1 2016 PURPOSE: UGT1A1*28/*6 as predictors of severe irinotecan-related diarrhea (SIRD) were duplicated by many studies. Irinotecan 46-56 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 9-15 27246726-1 2016 BACKGROUND: Patients with metastatic colorectal cancer whose disease has progressed on oxaliplatin- and irinotecan-containing regimens may benefit from EGFR-inhibiting monoclonal antibodies if they do not contain mutations in the KRAS gene (are "wild type"). Irinotecan 104-114 epidermal growth factor receptor Homo sapiens 152-156 26862009-0 2016 Impact of UGT1A1 genotype upon toxicities of combination with low-dose irinotecan plus platinum. Irinotecan 71-81 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 10-16 26862009-1 2016 AIM: Irinotecan-induced severe toxicities are possibly related to UGT1A1*6 and *28 genotypes. Irinotecan 5-15 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 66-72 26862009-2 2016 However, the correlation between UGT1A1 polymorphisms and the risk of toxicities induced by low-dose irinotecan plus platinum combination therapy still remains controversial. Irinotecan 101-111 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-39 26862009-3 2016 This prospective observational study aimed to examine the correlation between UGT1A1 genotypes and clinical outcomes of low-dose irinotecan (median 60 mg/m(2) , range 25-115 mg/m(2) ) plus platinum in Japanese patients with solid tumors. Irinotecan 129-139 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 78-84 26862009-10 2016 CONCLUSION: UGT1A1 genotype has a major impact on the increased risk of severe hematological toxicities, even in low-dose irinotecan regimens. Irinotecan 122-132 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-18 26862009-11 2016 UGT1A1 genotypes are useful biomarkers for predicting severe hematological toxicities in patients treated with irinotecan plus platinum analog. Irinotecan 111-121 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 27143148-1 2016 PURPOSE: Patients with unresectable wild-type KRAS metastatic colorectal cancer benefit from fluoropyrimidines (FP), oxaliplatin (O), irinotecan (I), bevacizumab (Bev), and epithelial growth factor receptor inhibitors (EGFRI). Irinotecan 134-144 KRAS proto-oncogene, GTPase Homo sapiens 46-50 27197307-0 2016 Irinotecan-Induced Gastrointestinal Dysfunction and Pain Are Mediated by Common TLR4-Dependent Mechanisms. Irinotecan 0-10 toll-like receptor 4 Mus musculus 80-84 27197307-9 2016 TLR4 deletion attenuated irinotecan-induced gut toxicity, with improvements in weight loss (P = 0.0003) and diarrhea (P < 0.0001). Irinotecan 25-35 toll-like receptor 4 Mus musculus 0-4 27197307-13 2016 These data indicate that TLR4 is uniquely positioned to mediate irinotecan-induced gut toxicity and pain, highlighting the possibility of a targetable gut/CNS axis for improved toxicity outcomes. Irinotecan 64-74 toll-like receptor 4 Mus musculus 25-29 27313739-16 2016 In conclusion, panitumumab and irinotecan as salvage therapy for mCRC KRAS wild-type patients with skin toxicity prevention exhibits limited efficacy. Irinotecan 31-41 KRAS proto-oncogene, GTPase Homo sapiens 70-74 27246726-1 2016 BACKGROUND: Patients with metastatic colorectal cancer whose disease has progressed on oxaliplatin- and irinotecan-containing regimens may benefit from EGFR-inhibiting monoclonal antibodies if they do not contain mutations in the KRAS gene (are "wild type"). Irinotecan 104-114 KRAS proto-oncogene, GTPase Homo sapiens 230-234 27380948-4 2016 RESULTS: Meta-analysis showed east-Asian patients expressing SLCO1B1 521T>C or 1118G>A to have a two- to fourfold increased risk of irinotecan-induced neutropenia but not diarrhea. Irinotecan 138-148 solute carrier organic anion transporter family member 1B1 Homo sapiens 61-68 27385990-0 2016 Prospective study of the UGT1A1*27 gene polymorphism during irinotecan therapy in patients with lung cancer: Results of Lung Oncology Group in Kyusyu (LOGIK1004B). Irinotecan 60-70 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 25-31 27385990-2 2016 To evaluate the effect of the UGT1A1*27 polymorphism in irinotecan therapy, we conducted a prospective study. Irinotecan 56-66 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 30-36 27385990-10 2016 In an evaluation of the side effects of irinotecan, patients with UGT1A1*28 and UGT1A1*6 polymorphisms had a higher tendency to experience febrile neutropenia than wild type (25% and 32% vs. 14%). Irinotecan 40-50 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 66-72 27385990-10 2016 In an evaluation of the side effects of irinotecan, patients with UGT1A1*28 and UGT1A1*6 polymorphisms had a higher tendency to experience febrile neutropenia than wild type (25% and 32% vs. 14%). Irinotecan 40-50 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 80-86 27340349-2 2016 In addition, the combined strategies of administering EGFR mAbs and traditional cytotoxic agents, such as 5-fluorouracil, oxaliplatin and irinotecan, have resulted in a more complicated management of CRC treatment-related side effects compared with EGFR mAb monotherapy. Irinotecan 138-148 epidermal growth factor receptor Homo sapiens 54-58 27340349-2 2016 In addition, the combined strategies of administering EGFR mAbs and traditional cytotoxic agents, such as 5-fluorouracil, oxaliplatin and irinotecan, have resulted in a more complicated management of CRC treatment-related side effects compared with EGFR mAb monotherapy. Irinotecan 138-148 epidermal growth factor receptor Homo sapiens 249-253 27009215-8 2016 In immunodeficient mice, systemically transplanted HB1.F3.CE stem cells migrated toward the tumor implanted by the TCCSUP bladder cancer cell line, and, in combination with CPT-11, the volume of tumors was significantly reduced. Irinotecan 173-179 paternally expressed 10 Mus musculus 51-54 27269636-7 2016 Meta-analysis showed an decreased risk of irinotecan-induced neutropenia in patients expressing ABCB1 2677G>T/G (odds ratio [OR]: 0.24; 95% CI: 0.1-0.59; p = 0.002) but increased risk for ABCC2 3972T>T (OR: 1.67; 95% CI: 1.01-2.74; p = 0.04). Irinotecan 42-52 ATP binding cassette subfamily B member 1 Homo sapiens 96-101 27269636-7 2016 Meta-analysis showed an decreased risk of irinotecan-induced neutropenia in patients expressing ABCB1 2677G>T/G (odds ratio [OR]: 0.24; 95% CI: 0.1-0.59; p = 0.002) but increased risk for ABCC2 3972T>T (OR: 1.67; 95% CI: 1.01-2.74; p = 0.04). Irinotecan 42-52 ATP binding cassette subfamily C member 2 Homo sapiens 191-196 27269636-8 2016 ABCG2 34G>A was associated with a threefold increased risk of irinotecan-induced diarrhea (95% CI: 1.00-6.24; p = 0.05). Irinotecan 65-75 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 0-5 26518357-11 2016 These findings suggest that the reduction in the serum SN-38 concentration following co-administration of ciprofloxacin during irinotecan treatment is due, at least partly, to the decreased enterohepatic circulation of SN-38 through the non-competitive inhibition of intestinal beta-glucuronidase-mediated SN-38-G deconjugation. Irinotecan 219-224 glucuronidase beta Homo sapiens 278-296 27070848-6 2016 The release of therapeutic drug SN-38 in the presence of CTB was confirmed by HPLC and prodrug 1a showed potent in vitro cytotoxicity against all tested cell lines. Irinotecan 32-37 cathepsin B Homo sapiens 57-60 27135737-5 2016 Upon preferential cancer cell delivery and uptake, aided by biotin, the enzyme-triggered theranostic prodrug 1 is cleaved by NQO1, with the subsequent release of SN-38, inhibiting topoisomerase I, leading to apoptosis. Irinotecan 162-167 NAD(P)H quinone dehydrogenase 1 Homo sapiens 125-129 27135737-6 2016 The drug release and induced apoptosis of cancer cells expressing both biotin receptors and high levels of NQO1 was simultaneously monitored via the innate fluorescence of the released SN-38 by confocal microscopy. Irinotecan 185-190 NAD(P)H quinone dehydrogenase 1 Homo sapiens 107-111 27070088-0 2016 The novel tankyrase inhibitor (AZ1366) enhances irinotecan activity in tumors that exhibit elevated tankyrase and irinotecan resistance. Irinotecan 48-58 tankyrase Homo sapiens 10-19 27070088-0 2016 The novel tankyrase inhibitor (AZ1366) enhances irinotecan activity in tumors that exhibit elevated tankyrase and irinotecan resistance. Irinotecan 48-58 tankyrase Homo sapiens 100-109 27070088-0 2016 The novel tankyrase inhibitor (AZ1366) enhances irinotecan activity in tumors that exhibit elevated tankyrase and irinotecan resistance. Irinotecan 114-124 tankyrase Homo sapiens 10-19 26518357-0 2016 The Inhibitory Effect of Ciprofloxacin on the beta-Glucuronidase-mediated Deconjugation of the Irinotecan Metabolite SN-38-G. Irinotecan 95-105 glucuronidase beta Homo sapiens 46-64 26518357-4 2016 In this study, we established an in vitro system to evaluate the beta-glucuronidase-mediated deconjugation of the irinotecan metabolite SN-38-G to its active SN-38 form and the effect of ciprofloxacin thereon. Irinotecan 114-124 glucuronidase beta Homo sapiens 65-83 26518357-4 2016 In this study, we established an in vitro system to evaluate the beta-glucuronidase-mediated deconjugation of the irinotecan metabolite SN-38-G to its active SN-38 form and the effect of ciprofloxacin thereon. Irinotecan 136-141 glucuronidase beta Homo sapiens 65-83 26518357-4 2016 In this study, we established an in vitro system to evaluate the beta-glucuronidase-mediated deconjugation of the irinotecan metabolite SN-38-G to its active SN-38 form and the effect of ciprofloxacin thereon. Irinotecan 158-163 glucuronidase beta Homo sapiens 65-83 26518357-7 2016 Ciprofloxacin and phenolphthalein-beta-D-glucuronide (PhePG), a typical beta-glucuronidase substrate, significantly decreased SN-38-G deconjugation and, hence SN-38 formation. Irinotecan 126-131 glucuronidase beta Homo sapiens 72-90 26518357-9 2016 Ciprofloxacin showed a dose-dependent inhibitory effect on the beta-glucuronidase-mediated conversion of SN-38-G to SN-38 with a half-maximal inhibitory concentration (IC50 ) value of 83.8 muM. Irinotecan 105-110 glucuronidase beta Homo sapiens 63-81 26518357-11 2016 These findings suggest that the reduction in the serum SN-38 concentration following co-administration of ciprofloxacin during irinotecan treatment is due, at least partly, to the decreased enterohepatic circulation of SN-38 through the non-competitive inhibition of intestinal beta-glucuronidase-mediated SN-38-G deconjugation. Irinotecan 55-60 glucuronidase beta Homo sapiens 278-296 26518357-11 2016 These findings suggest that the reduction in the serum SN-38 concentration following co-administration of ciprofloxacin during irinotecan treatment is due, at least partly, to the decreased enterohepatic circulation of SN-38 through the non-competitive inhibition of intestinal beta-glucuronidase-mediated SN-38-G deconjugation. Irinotecan 219-224 glucuronidase beta Homo sapiens 278-296 26115978-1 2016 OBJECTIVE: This study sought to evaluate the toxicity and curative effect of irinotecan plus cisplatin neoadjuvant chemotherapy (NACT) for stage Ib2, IIa2, and IIb cervical cancer patients. Irinotecan 77-87 mitogen-activated protein kinase 8 interacting protein 2 Homo sapiens 145-148 27037412-9 2016 In combination with irinotecan or doxorubicin, FAP-DR5 treatment resulted in substantial tumor regression in patient-derived xenograft models. Irinotecan 20-30 fibroblast activation protein alpha Homo sapiens 47-50 25917796-8 2016 UbcH10 suppression decreased CRC cell growth rate (at least in part through deregulation of Cyclin B and ERK1) and sensitized them to pharmacological treatments with irinotecan, SN-38 and cetuximab (at least in part through a down-regulation of AKT). Irinotecan 166-176 ubiquitin conjugating enzyme E2 C Homo sapiens 0-6 25917796-8 2016 UbcH10 suppression decreased CRC cell growth rate (at least in part through deregulation of Cyclin B and ERK1) and sensitized them to pharmacological treatments with irinotecan, SN-38 and cetuximab (at least in part through a down-regulation of AKT). Irinotecan 178-183 ubiquitin conjugating enzyme E2 C Homo sapiens 0-6 26826119-8 2016 Treatment of the Gb3-expressing gastric carcinoma cell line St3051 with STxB coupled to SN38, the active metabolite of the topoisomerase type I inhibitor irinotecan, resulted in >100-fold increased cytotoxicity, as compared with irinotecan alone. Irinotecan 154-164 alpha 1,4-galactosyltransferase (P blood group) Homo sapiens 17-20 26826119-8 2016 Treatment of the Gb3-expressing gastric carcinoma cell line St3051 with STxB coupled to SN38, the active metabolite of the topoisomerase type I inhibitor irinotecan, resulted in >100-fold increased cytotoxicity, as compared with irinotecan alone. Irinotecan 232-242 alpha 1,4-galactosyltransferase (P blood group) Homo sapiens 17-20 27037412-9 2016 In combination with irinotecan or doxorubicin, FAP-DR5 treatment resulted in substantial tumor regression in patient-derived xenograft models. Irinotecan 20-30 TNF receptor superfamily member 10b Homo sapiens 51-54 26988911-1 2016 We previously reported that ATP7B is involved in cisplatin resistance and ATP7A confers multidrug resistance (MDR) in cancer cells.In this study, we show that ATP7B expressing cells also are resistant to doxorubicin, SN-38, etoposide, and paclitaxel as well as cisplatin.In ATP7B expressing cells, doxorubicin relocated from the nuclei to the late-endosome at 4 hours after doxorubicin exposure. Irinotecan 217-222 ATPase, Cu++ transporting, alpha polypeptide Gallus gallus 74-79 26987941-11 2016 Irinotecan also induced increased ROS production in enterocytes, oxidative stress, IL-1beta production, neutrophil and eosinophil influx in the ileum. Irinotecan 0-10 interleukin 1 beta Mus musculus 83-91 26581898-6 2016 Irinotecan also caused a decrease in TLR4 and 5, and an increase in GFAP expression in jejuna crypt at 96 and 120 h (all P < 0.05); with lower TLR4 expression associated with lower pain (P = 0.012). Irinotecan 0-10 toll-like receptor 4 Rattus norvegicus 37-47 26581898-6 2016 Irinotecan also caused a decrease in TLR4 and 5, and an increase in GFAP expression in jejuna crypt at 96 and 120 h (all P < 0.05); with lower TLR4 expression associated with lower pain (P = 0.012). Irinotecan 0-10 glial fibrillary acidic protein Rattus norvegicus 68-72 26581898-6 2016 Irinotecan also caused a decrease in TLR4 and 5, and an increase in GFAP expression in jejuna crypt at 96 and 120 h (all P < 0.05); with lower TLR4 expression associated with lower pain (P = 0.012). Irinotecan 0-10 toll-like receptor 4 Rattus norvegicus 37-41 27122675-1 2016 AIM: To examine the predictive effects of baseline serum bilirubin levels and UDP-glucuronosyltransferase (UGT) 1A1*28 polymorphism on response of colorectal cancer to irinotecan-based chemotherapy. Irinotecan 168-178 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 78-115 27122675-16 2016 CONCLUSION: CoBil and UGT1A1*28 are both independent biomarkers for predicting the treatment response of mCRC patients to irinotecan-based chemotherapy. Irinotecan 122-132 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 22-28 26988911-1 2016 We previously reported that ATP7B is involved in cisplatin resistance and ATP7A confers multidrug resistance (MDR) in cancer cells.In this study, we show that ATP7B expressing cells also are resistant to doxorubicin, SN-38, etoposide, and paclitaxel as well as cisplatin.In ATP7B expressing cells, doxorubicin relocated from the nuclei to the late-endosome at 4 hours after doxorubicin exposure. Irinotecan 217-222 ATPase copper transporting beta Gallus gallus 159-164 26988911-1 2016 We previously reported that ATP7B is involved in cisplatin resistance and ATP7A confers multidrug resistance (MDR) in cancer cells.In this study, we show that ATP7B expressing cells also are resistant to doxorubicin, SN-38, etoposide, and paclitaxel as well as cisplatin.In ATP7B expressing cells, doxorubicin relocated from the nuclei to the late-endosome at 4 hours after doxorubicin exposure. Irinotecan 217-222 ATPase copper transporting beta Gallus gallus 159-164 27091477-0 2016 Epidermal growth factor receptor inhibitor with fluorouracil, leucovorin, and irinotecan as an alternative treatment for advanced upper tract urothelial carcinoma: a case report. Irinotecan 78-88 epidermal growth factor receptor Homo sapiens 0-32 26900660-1 2016 Human carboxylesterase 2 (hCE2), one of the major carboxylesterases in the human intestine and various tumour tissues, plays important roles in the oral bioavailability and treatment outcomes of ester- or amide-containing drugs or prodrugs, such as anticancer agents CPT-11 (irinotecan) and LY2334737 (gemcitabine). Irinotecan 267-273 carboxylesterase 2 Homo sapiens 6-24 26900660-1 2016 Human carboxylesterase 2 (hCE2), one of the major carboxylesterases in the human intestine and various tumour tissues, plays important roles in the oral bioavailability and treatment outcomes of ester- or amide-containing drugs or prodrugs, such as anticancer agents CPT-11 (irinotecan) and LY2334737 (gemcitabine). Irinotecan 267-273 carboxylesterase 2 Homo sapiens 26-30 26900660-1 2016 Human carboxylesterase 2 (hCE2), one of the major carboxylesterases in the human intestine and various tumour tissues, plays important roles in the oral bioavailability and treatment outcomes of ester- or amide-containing drugs or prodrugs, such as anticancer agents CPT-11 (irinotecan) and LY2334737 (gemcitabine). Irinotecan 267-273 carboxylesterase 1 Homo sapiens 50-67 26900660-1 2016 Human carboxylesterase 2 (hCE2), one of the major carboxylesterases in the human intestine and various tumour tissues, plays important roles in the oral bioavailability and treatment outcomes of ester- or amide-containing drugs or prodrugs, such as anticancer agents CPT-11 (irinotecan) and LY2334737 (gemcitabine). Irinotecan 275-285 carboxylesterase 2 Homo sapiens 6-24 26900660-1 2016 Human carboxylesterase 2 (hCE2), one of the major carboxylesterases in the human intestine and various tumour tissues, plays important roles in the oral bioavailability and treatment outcomes of ester- or amide-containing drugs or prodrugs, such as anticancer agents CPT-11 (irinotecan) and LY2334737 (gemcitabine). Irinotecan 275-285 carboxylesterase 2 Homo sapiens 26-30 26900660-1 2016 Human carboxylesterase 2 (hCE2), one of the major carboxylesterases in the human intestine and various tumour tissues, plays important roles in the oral bioavailability and treatment outcomes of ester- or amide-containing drugs or prodrugs, such as anticancer agents CPT-11 (irinotecan) and LY2334737 (gemcitabine). Irinotecan 275-285 carboxylesterase 1 Homo sapiens 50-67 27461651-2 2016 DPYD*2A/*5A/*9A and UGT1A1 * 6/*28 polymorphisms are related to the toxicity of fluorouracil drugs and irinotecan, respectively. Irinotecan 103-113 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 20-26 26896610-5 2016 The SN-38/USPIO-loaded siRNA-PEG mixed micelleplexes passively targeted to tumor regions and synergistically facilitated VEGF silencing and chemotherapy, thus efficiently suppressing tumor growth via a multi-dose therapy regimen. Irinotecan 4-9 vascular endothelial growth factor A Homo sapiens 121-125 26857987-3 2016 Etirinotecan pegol, comprising irinotecan bound to polyethylene glycol by a biodegradable linker, is a new cytotoxic agent for patients with MBC that has achieved encouraging response rates in phase II studies; it has been further evaluated in the phase III BEACON trial. Irinotecan 2-12 ubiquitin like 5 Homo sapiens 258-264 27034809-6 2016 The presence of the uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) *28 polymorphism is associated with toxicity from irinotecan, although it has not been universally applied. Irinotecan 127-137 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 20-67 27034809-6 2016 The presence of the uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) *28 polymorphism is associated with toxicity from irinotecan, although it has not been universally applied. Irinotecan 127-137 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 69-75 26719532-4 2016 However, the full potential of irinotecan treatment is hindered by several cancer cell survival mechanisms, including ATP-binding cassette G2 (ABCG2) transporter-mediated irinotecan efflux from cells. Irinotecan 31-41 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 118-141 27080842-6 2016 Other tests are also commonly performed to address the toxicity of certain anticancer drugs (DPYD-capecitabine, UGT1A1-irinotecan, TPMT 6-mercaptopurine). Irinotecan 119-129 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 112-118 26872382-3 2016 We found that adenovirus prevents Chk1-mediated checkpoint activation through inactivation of Mre11 and downregulation of the pChk1 adaptor-protein, Claspin, in cells with high levels of DNA-damage induced by the cytotoxic drugs gemcitabine and irinotecan. Irinotecan 245-255 checkpoint kinase 1 Homo sapiens 34-38 26872382-3 2016 We found that adenovirus prevents Chk1-mediated checkpoint activation through inactivation of Mre11 and downregulation of the pChk1 adaptor-protein, Claspin, in cells with high levels of DNA-damage induced by the cytotoxic drugs gemcitabine and irinotecan. Irinotecan 245-255 claspin Homo sapiens 149-156 26719532-4 2016 However, the full potential of irinotecan treatment is hindered by several cancer cell survival mechanisms, including ATP-binding cassette G2 (ABCG2) transporter-mediated irinotecan efflux from cells. Irinotecan 31-41 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 143-148 26719532-4 2016 However, the full potential of irinotecan treatment is hindered by several cancer cell survival mechanisms, including ATP-binding cassette G2 (ABCG2) transporter-mediated irinotecan efflux from cells. Irinotecan 171-181 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 118-141 26719532-4 2016 However, the full potential of irinotecan treatment is hindered by several cancer cell survival mechanisms, including ATP-binding cassette G2 (ABCG2) transporter-mediated irinotecan efflux from cells. Irinotecan 171-181 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 143-148 26719532-5 2016 Here, we demonstrate that benzoporphyrin derivative-based photodynamic therapy (PDT), a photochemical cytotoxic modality that activates the apoptotic pathway, reduced ABCG2 expression to increase intracellular irinotecan levels in pancreatic cancer. Irinotecan 210-220 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 167-172 26719532-7 2016 Although PDT potentiated irinotecan treatment, we also demonstrate that irinotecan reduced the tumoral expression of monocarboxylate transporter 4, which was upregulated by PDT. Irinotecan 72-82 solute carrier family 16 member 3 Homo sapiens 117-146 26857783-4 2016 RESULTS AND CONCLUSION: UGT1A1*6 and UGT1A1*28 polymorphisms were associated with severe neutropenia (p = 0.025, p = 0.022, respectively) but not diarrhea (p = 0.343, p = 0.185, respectively), and ABCB1- 3435C>T polymorphism was not associated with irinotecan induced severe toxicities (p = 0.457, p = 0.161, respectively). Irinotecan 252-262 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 24-30 26857783-4 2016 RESULTS AND CONCLUSION: UGT1A1*6 and UGT1A1*28 polymorphisms were associated with severe neutropenia (p = 0.025, p = 0.022, respectively) but not diarrhea (p = 0.343, p = 0.185, respectively), and ABCB1- 3435C>T polymorphism was not associated with irinotecan induced severe toxicities (p = 0.457, p = 0.161, respectively). Irinotecan 252-262 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 37-43 26773202-1 2016 Certain genetic polymorphisms of UDP glucuronosyltransferase 1 family, polypeptide A1 (UGT1A1) can reduce gene expression (*28, *60, *93) or activity (*6), thereby altering the pharmacokinetics, pharmacodynamics, and the risk of toxicities of UGT1A1 substrates, of which irinotecan is a widely-described example. Irinotecan 271-281 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-85 26773202-1 2016 Certain genetic polymorphisms of UDP glucuronosyltransferase 1 family, polypeptide A1 (UGT1A1) can reduce gene expression (*28, *60, *93) or activity (*6), thereby altering the pharmacokinetics, pharmacodynamics, and the risk of toxicities of UGT1A1 substrates, of which irinotecan is a widely-described example. Irinotecan 271-281 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 87-93 26773202-1 2016 Certain genetic polymorphisms of UDP glucuronosyltransferase 1 family, polypeptide A1 (UGT1A1) can reduce gene expression (*28, *60, *93) or activity (*6), thereby altering the pharmacokinetics, pharmacodynamics, and the risk of toxicities of UGT1A1 substrates, of which irinotecan is a widely-described example. Irinotecan 271-281 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 243-249 26541586-4 2016 This antibody-drug conjugate comprises the active metabolite of irinotecan, SN-38, conjugated to an anti-Trop-2 antibody. Irinotecan 64-74 tumor associated calcium signal transducer 2 Homo sapiens 105-111 26531131-9 2016 As GATA6 is a transcription factor that controls expression of multiple genes regulating peritoneal B-1 cell development and translocation, elimination of GATA6(+) LPMs led to a great reduction in B-1 cells in the peritoneal cavity after CPT-11 treatment. Irinotecan 238-244 GATA binding protein 6 Mus musculus 3-8 26531131-9 2016 As GATA6 is a transcription factor that controls expression of multiple genes regulating peritoneal B-1 cell development and translocation, elimination of GATA6(+) LPMs led to a great reduction in B-1 cells in the peritoneal cavity after CPT-11 treatment. Irinotecan 238-244 GATA binding protein 6 Mus musculus 155-160 26381420-3 2016 Cetuximab is a monoclonal antibody against EGFR with additive preclinical activity with irinotecan. Irinotecan 88-98 epidermal growth factor receptor Homo sapiens 43-47 26531131-7 2016 Consistent with this, the transcription factor GATA6 specifically expressed in LPMs was barely detectable in the macrophages from CPT-11-treated mice, indicative of elimination of LPMs. Irinotecan 130-136 GATA binding protein 6 Mus musculus 47-52 26541586-4 2016 This antibody-drug conjugate comprises the active metabolite of irinotecan, SN-38, conjugated to an anti-Trop-2 antibody. Irinotecan 76-81 tumor associated calcium signal transducer 2 Homo sapiens 105-111 26284333-9 2016 EREG expression appears as an independent prognostic marker in patients with mCRC receiving first-line irinotecan-based chemotherapy. Irinotecan 103-113 epiregulin Homo sapiens 0-4 27057810-5 2016 This changed markedly over the last decade with the approval of irinotecan, oxaliplatin, capecitabine, humanized monoclonal antibodies that target either vascular endothelial growth factor (bevacizumab, aflibercept, and ramucirumab) or the epidermal growth factor receptor (cetuximab and panitumumab), and, most recently, regorafenib and trifluridine/tipiracil. Irinotecan 64-74 vascular endothelial growth factor A Homo sapiens 154-188 26830078-0 2016 Correlation of UGT1A1(*)28 and (*)6 polymorphisms with irinotecan-induced neutropenia in Thai colorectal cancer patients. Irinotecan 55-65 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 15-21 26830078-1 2016 UDP-glucuronosyltransferase1A1 (UGT1A1) polymorphisms have been related with irinotecan toxicity. Irinotecan 77-87 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-30 26830078-1 2016 UDP-glucuronosyltransferase1A1 (UGT1A1) polymorphisms have been related with irinotecan toxicity. Irinotecan 77-87 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 32-38 26830078-2 2016 The purpose of this study was to determine the associations between UGT1A1(*)28 and (*)6 polymorphisms and irinotecan toxicity in Thai patients with metastatic colorectal cancer. Irinotecan 107-117 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 68-74 26830078-10 2016 Both UGT1A1(*)28 and (*)6 polymorphisms may have an increased risk of irinotecan-induced neutropenia in Thai colorectal cancer patients. Irinotecan 70-80 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 5-11 25869015-5 2016 For irinotecan area under the concentration-time curve (AUC0-24), we replicated ABCC2 -24C>T; however, ABCC2 -24C>T only predicted a small fraction of the variance. Irinotecan 4-14 ATP binding cassette subfamily C member 2 Homo sapiens 80-85 25869015-6 2016 For SN-38 AUC0-24 and the glucuronidation ratio, we replicated UGT1A1*28 and UGT1A1*93. Irinotecan 4-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 63-69 25869015-6 2016 For SN-38 AUC0-24 and the glucuronidation ratio, we replicated UGT1A1*28 and UGT1A1*93. Irinotecan 4-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 77-83 25869015-7 2016 In addition to UGT1A1*28, this study independently validated UGT1A1*93 and SLCO1B1*1b as new predictors of irinotecan neutropenia. Irinotecan 107-117 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 61-67 25869015-7 2016 In addition to UGT1A1*28, this study independently validated UGT1A1*93 and SLCO1B1*1b as new predictors of irinotecan neutropenia. Irinotecan 107-117 solute carrier organic anion transporter family member 1B1 Homo sapiens 75-82 26551156-9 2016 Finally, cetuximab (moAb) and irinotecan (chemotherapy) drugs are analyzed as first-line treatment of colorectal cancer with few KRAS mutated cells. Irinotecan 30-40 KRAS proto-oncogene, GTPase Homo sapiens 129-133 26820647-0 2016 Molecular Genetics External Quality Assessment Pilot Scheme for Irinotecan-Related UGT1A1 Genotyping in China. Irinotecan 64-74 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 83-89 26820647-2 2016 Molecular testing for UGT1A1 genotyping is increasingly required in China for optimum irinotecan administration. Irinotecan 86-96 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 22-28 26811156-0 2016 Prospective analysis of UGT1A1 promoter polymorphism for irinotecan dose escalation in metastatic colorectal cancer patients treated with bevacizumab plus FOLFIRI as the first-line setting: study protocol for a randomized controlled trial. Irinotecan 57-67 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 24-30 26811156-3 2016 Therefore, we plan to explore the effect of the genetic polymorphism of uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) for irinotecan detoxification in mCRC patients. Irinotecan 133-143 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 72-119 26811156-3 2016 Therefore, we plan to explore the effect of the genetic polymorphism of uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) for irinotecan detoxification in mCRC patients. Irinotecan 133-143 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 121-127 26811156-6 2016 METHODS/DESIGN: This trial is a prospective, multicenter, randomized clinical trial comparing UGT1A1 promoter polymorphism for irinotecan dose escalation in mCRC patients administered with bevacizumab plus FOLFIRI as the first-line setting. Irinotecan 127-137 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 94-100 26811156-8 2016 The study group receive a biweekly FOLFIRI regimen, with irinotecan dose escalation based on UGT1A1 genotyping; whereas the control group receive the conventional biweekly FOLFIRI regimen without UGT1A1 genotyping. Irinotecan 57-67 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-99 26811156-10 2016 DISCUSSION: Patients with mCRC undergoing UGT1A1 genotyping may receive escalated doses of irinotecan for a potentially more favorable clinical response and outcome, in addition to comparable toxicities. Irinotecan 91-101 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 42-48 26808671-0 2016 Phenotyping of UGT1A1 Activity Using Raltegravir Predicts Pharmacokinetics and Toxicity of Irinotecan in FOLFIRI. Irinotecan 91-101 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 15-21 26808671-1 2016 BACKGROUND: Irinotecan toxicity correlates with UGT1A1 activity. Irinotecan 12-22 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 48-54 26801902-8 2016 The gene and protein expression of ABCG2/BCRP was up-regulated in the resistant sub-lines and functional assays revealed BCRP as a key mediator of SN-38 resistance. Irinotecan 147-152 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 35-40 26801902-8 2016 The gene and protein expression of ABCG2/BCRP was up-regulated in the resistant sub-lines and functional assays revealed BCRP as a key mediator of SN-38 resistance. Irinotecan 147-152 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 41-45 26801902-8 2016 The gene and protein expression of ABCG2/BCRP was up-regulated in the resistant sub-lines and functional assays revealed BCRP as a key mediator of SN-38 resistance. Irinotecan 147-152 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 121-125 26801902-9 2016 CONCLUSIONS: Based on our preclinical results, we suggest analyzing the predictive value of the BCRP in breast cancer patients scheduled for irinotecan treatment. Irinotecan 141-151 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 96-100 26752698-7 2016 Taken together, our data demonstrated for the first time that the combination of irinotecan and gefitinib showed potential benefit in HCC, which suggests that Rad51 is a promising target and provides a rationale for clinical trials investigating the efficacy of the combination of topoisomerase I inhibitors and gefitinib in HCC. Irinotecan 81-91 RAD51 recombinase Homo sapiens 159-164 26706406-0 2016 Irinotecan (CPT-11)-induced elevation of bile acids potentiates suppression of IL-10 expression. Irinotecan 0-10 interleukin 10 Mus musculus 79-84 26706406-0 2016 Irinotecan (CPT-11)-induced elevation of bile acids potentiates suppression of IL-10 expression. Irinotecan 12-18 interleukin 10 Mus musculus 79-84 26706406-5 2016 Analysis of immune cell populations further showed that CPT-11 treatment significantly decreased the IL-10-producing CD4 T cell frequency in intestinal lamina propria lymphocytes, but not in spleen or mesenteric lymph nodes. Irinotecan 56-62 interleukin 10 Mus musculus 101-106 26706406-6 2016 In vitro cell culture studies showed that the addition of bile acids deoxycholic acid and taurodeoxycholic acid accelerated the CPT-11-induced suppression of IL-10 secretion by activated CD4(+) naive T cells isolated from mouse splenocytes. Irinotecan 128-134 interleukin 10 Mus musculus 158-163 26706406-7 2016 These results showed that CPT-11 treatment caused metabolic changes in the composition of bile acids that altered CPT-11-induced suppression of IL-10 expression. Irinotecan 26-32 interleukin 10 Mus musculus 144-149 26706406-7 2016 These results showed that CPT-11 treatment caused metabolic changes in the composition of bile acids that altered CPT-11-induced suppression of IL-10 expression. Irinotecan 114-120 interleukin 10 Mus musculus 144-149 26098842-2 2016 This study evaluates the cost-effectiveness of UGT1A1 genotyping in patients with metastatic colorectal cancer undergoing irinotecan-based chemotherapy compared to no testing from the perspective of the German statutory health insurance. Irinotecan 122-132 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 47-53 26872008-0 2016 Phase II and UGT1A1 Polymorphism Study of Two Different Irinotecan Dosages Combined with Cisplatin as First-Line Therapy for Advanced Gastric Cancer. Irinotecan 56-66 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 13-19 26577300-1 2016 A phase II study indicates that sacituzumab govitecan (IMMU-132), a Trop-2-specific antibody linked to the irinotecan metabolite SN-38, prolongs the progression-free survival of patients with advanced triple-negative breast cancer. Irinotecan 107-117 tumor associated calcium signal transducer 2 Homo sapiens 68-74 26577300-1 2016 A phase II study indicates that sacituzumab govitecan (IMMU-132), a Trop-2-specific antibody linked to the irinotecan metabolite SN-38, prolongs the progression-free survival of patients with advanced triple-negative breast cancer. Irinotecan 129-134 tumor associated calcium signal transducer 2 Homo sapiens 68-74 29787016-8 2016 Postoperatively, the patient received six courses of combined therapy with irinotecan and cisplatin (CPT-P therapy), and the patient has survived disease- free for over two years. Irinotecan 75-85 ceramide-1-phosphate transfer protein Homo sapiens 101-106 26728136-0 2016 PEO-PPO-PEO/Poly(DL-lactide-co-caprolactone) Nanoparticles as Carriers for SN-38: Design, Optimization and Nano-Bio Interface Interactions. Irinotecan 75-80 protoporphyrinogen oxidase Homo sapiens 4-7 26728136-4 2016 In this article we defined the optimal region for manufacturing of SN-38 loaded PEO-PPO-PEO/P(DL)LCL nanoparticles (NPs) with high efficacy of encapsulation, suitable particle size and different surface properties, using D-optimal design and nanoprecipitation as production method. Irinotecan 67-72 protoporphyrinogen oxidase Homo sapiens 84-87 26526067-4 2016 Based on the fact that liver-specific organic anion transporting polypeptide (OATP)1B1, a member of the same family as OATP2B1, is known to be involved in hepatic transport of SN-38, we hypothesized that intestinal transporter OATP2B1 contributes to the accumulation of SN-38 in gastrointestinal tissues, and its inhibition would help prevent associated toxicity. Irinotecan 176-181 solute carrier organic anion transporter family member 2B1 L homeolog Xenopus laevis 119-126 26526067-4 2016 Based on the fact that liver-specific organic anion transporting polypeptide (OATP)1B1, a member of the same family as OATP2B1, is known to be involved in hepatic transport of SN-38, we hypothesized that intestinal transporter OATP2B1 contributes to the accumulation of SN-38 in gastrointestinal tissues, and its inhibition would help prevent associated toxicity. Irinotecan 176-181 solute carrier organic anion transporter family member 2B1 L homeolog Xenopus laevis 227-234 26526067-4 2016 Based on the fact that liver-specific organic anion transporting polypeptide (OATP)1B1, a member of the same family as OATP2B1, is known to be involved in hepatic transport of SN-38, we hypothesized that intestinal transporter OATP2B1 contributes to the accumulation of SN-38 in gastrointestinal tissues, and its inhibition would help prevent associated toxicity. Irinotecan 270-275 solute carrier organic anion transporter family member 2B1 L homeolog Xenopus laevis 119-126 26526067-5 2016 We found that uptake of SN-38 by OATP2B1-expressing Xenopus oocytes was significantly higher than that by control oocytes. Irinotecan 24-29 solute carrier organic anion transporter family member 2B1 L homeolog Xenopus laevis 33-40 26526067-6 2016 OATP2B1-mediated uptake of SN-38 was saturable, pH dependent, and decreased in the presence of baicalin, cefixime, or fruit juices such as apple juice. Irinotecan 27-32 solute carrier organic anion transporter family member 2B1 L homeolog Xenopus laevis 0-7 26526067-8 2016 Thus, OATP2B1 contributes to the uptake of SN-38 by intestinal tissues, triggering gastrointestinal toxicity. Irinotecan 43-48 solute carrier organic anion transporter family member 2B1 L homeolog Xenopus laevis 6-13 25545261-1 2016 BACKGROUND: UGT1A1 is a polymorphic enzyme that has been associated with irinotecan drug metabolisms. Irinotecan 73-83 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-18 26966430-6 2016 The IC50 values of simvastatin alone and irinotecan alone were 115.4 +- 0.14 muM (r = 0.98) and 62.5 +- 0.18 muM (r = 0.98) in HT-29 cells without resistance to irinotecan. Irinotecan 41-51 latexin Homo sapiens 77-80 26966430-6 2016 The IC50 values of simvastatin alone and irinotecan alone were 115.4 +- 0.14 muM (r = 0.98) and 62.5 +- 0.18 muM (r = 0.98) in HT-29 cells without resistance to irinotecan. Irinotecan 41-51 latexin Homo sapiens 109-112 27072236-14 2016 Furthermore, as 5.FU and irinotecan metabolism is complex, numerous genes in addition to DPD and UGT1A1 should be investigated. Irinotecan 25-35 dihydropyrimidine dehydrogenase Homo sapiens 89-92 27072236-14 2016 Furthermore, as 5.FU and irinotecan metabolism is complex, numerous genes in addition to DPD and UGT1A1 should be investigated. Irinotecan 25-35 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 97-103 27229742-3 2016 METHODS: Subjects were mCRC patients with mutated KRAS who showed disease aggravation even after two regimens with oxaliplatin and irinotecan. Irinotecan 131-141 KRAS proto-oncogene, GTPase Homo sapiens 50-54 27229742-12 2016 CONCLUSIONS: The results suggest that third-line chemotherapy with combined bevacizumab and S-1 is safe and may delay the progression of mCRC resistant to oxaliplatin and irinotecan with mutated KRAS. Irinotecan 171-181 proteasome 26S subunit, non-ATPase 1 Homo sapiens 92-95 27843533-9 2016 Accordingly, NPOA could be a candidate chemosensitizer of CPT derivative agents such as irinotecan or topotecan in the future. Irinotecan 88-98 choline phosphotransferase 1 Homo sapiens 58-61 27803477-5 2016 1) In 2005, information regarding the genetic polymorphism of UGT1A1*28 was described in the package insert of the drug irinotecan in the United States. Irinotecan 120-130 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 62-68 27803477-7 2016 Through clinical research, we demonstrated that UGT1A1*6 was a significant factor for neutropenia, as induced by irinotecan. Irinotecan 113-123 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 48-54 26439663-3 2015 In this study, three kinds of linkers were designed to connect SN-38, the bioactive metabolite of the anticancer drug irinotecan (CPT-11), which is 100-1000 times more potent than CPT-11, with the anti-HER2 antibody trastuzumab to prepare three different ADC conjugates (T-SN38 A, B and C). Irinotecan 63-68 erb-b2 receptor tyrosine kinase 2 Homo sapiens 202-206 26439663-3 2015 In this study, three kinds of linkers were designed to connect SN-38, the bioactive metabolite of the anticancer drug irinotecan (CPT-11), which is 100-1000 times more potent than CPT-11, with the anti-HER2 antibody trastuzumab to prepare three different ADC conjugates (T-SN38 A, B and C). Irinotecan 118-128 erb-b2 receptor tyrosine kinase 2 Homo sapiens 202-206 26439663-3 2015 In this study, three kinds of linkers were designed to connect SN-38, the bioactive metabolite of the anticancer drug irinotecan (CPT-11), which is 100-1000 times more potent than CPT-11, with the anti-HER2 antibody trastuzumab to prepare three different ADC conjugates (T-SN38 A, B and C). Irinotecan 130-136 erb-b2 receptor tyrosine kinase 2 Homo sapiens 202-206 26664141-0 2015 The relationship between UGT1A1 gene polymorphism and irinotecan effect on extensive-stage small-cell lung cancer. Irinotecan 54-64 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 25-31 26485768-0 2015 Akt inhibition improves irinotecan treatment and prevents cell emergence by switching the senescence response to apoptosis. Irinotecan 24-34 AKT serine/threonine kinase 1 Homo sapiens 0-3 25915850-7 2015 Moreover, administration of DSB-inducing chemicals, camptothecin (CPT) or irinotecan, to Myc-ELAS1-expressing U2OS cells also induced efficient apoptosis with only 1/100th (CPT) or 1/5th (irinotecan) of the amounts of drugs required for this effect in Myc-vector-expressing cells. Irinotecan 74-84 MYC proto-oncogene, bHLH transcription factor Homo sapiens 89-92 25915850-7 2015 Moreover, administration of DSB-inducing chemicals, camptothecin (CPT) or irinotecan, to Myc-ELAS1-expressing U2OS cells also induced efficient apoptosis with only 1/100th (CPT) or 1/5th (irinotecan) of the amounts of drugs required for this effect in Myc-vector-expressing cells. Irinotecan 74-84 MYC proto-oncogene, bHLH transcription factor Homo sapiens 252-255 25915850-7 2015 Moreover, administration of DSB-inducing chemicals, camptothecin (CPT) or irinotecan, to Myc-ELAS1-expressing U2OS cells also induced efficient apoptosis with only 1/100th (CPT) or 1/5th (irinotecan) of the amounts of drugs required for this effect in Myc-vector-expressing cells. Irinotecan 188-198 MYC proto-oncogene, bHLH transcription factor Homo sapiens 89-92 26664141-11 2015 CONCLUSION: For irinotecan-based regimens in E-SCLC, the UGT1A1*28 and UGT1A1*6 locus mutations can be regarded as predictors for severe adverse events. Irinotecan 16-26 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 57-63 26664141-11 2015 CONCLUSION: For irinotecan-based regimens in E-SCLC, the UGT1A1*28 and UGT1A1*6 locus mutations can be regarded as predictors for severe adverse events. Irinotecan 16-26 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 71-77 25799948-3 2015 TACE with 100 mg of Irinotecan loaded into 2-ml of 70-150 microm drug-eluting beads was administrated every 4 weeks in patients with unilobar disease (2 treatments) and every 2 weeks in patients with bilobar disease (4 treatments). Irinotecan 20-30 ADAM metallopeptidase domain 17 Homo sapiens 0-4 27551256-4 2015 The decoy vascular endothelial growth factor receptor aflibercept has been approved in combination with 5-fluorouracil, leucovorin and irinotecan-based chemotherapy in metastatic colorectal cancer patients whose disease has progressed on a prior oxaliplatin-based chemotherapy regimen. Irinotecan 135-145 vascular endothelial growth factor A Homo sapiens 10-44 25216634-2 2015 Irinotecan-induced diarrhea is closely related with the UDP-glucuronosyltransferase 1A1-catalyzed glucuronidation of SN-38 which has been widely regarded to be the toxic substance basis of irinotecan. Irinotecan 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 56-87 25216634-2 2015 Irinotecan-induced diarrhea is closely related with the UDP-glucuronosyltransferase 1A1-catalyzed glucuronidation of SN-38 which has been widely regarded to be the toxic substance basis of irinotecan. Irinotecan 117-122 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 56-87 25216634-2 2015 Irinotecan-induced diarrhea is closely related with the UDP-glucuronosyltransferase 1A1-catalyzed glucuronidation of SN-38 which has been widely regarded to be the toxic substance basis of irinotecan. Irinotecan 189-199 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 56-87 26494856-2 2015 UDP-glucuronosyltransferase: (UGT1A1) polymorphism may be predictive of toxicity and efficacy of irinotecan. Irinotecan 97-107 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 30-36 26438158-3 2015 Here, we report that PARP inhibitors synergize with temozolomide (TMZ) or SN-38 to induce apoptosis and also somewhat enhance the cytotoxicity of doxorubicin, etoposide, or ifosfamide, whereas actinomycin D and vincristine show little synergism. Irinotecan 74-79 poly(ADP-ribose) polymerase 1 Homo sapiens 21-25 26637886-2 2015 We conducted a multicenter phase II study of nedaplatin (NP) and irinotecan (CPT) for SCC of the lung. Irinotecan 65-75 serpin family B member 3 Homo sapiens 86-89 26692528-0 2015 Clinical Implication of UGT1A1 Promoter Polymorphism for Irinotecan Dose Escalation in Metastatic Colorectal Cancer Patients Treated with Bevacizumab Combined with FOLFIRI in the First-line Setting. Irinotecan 57-67 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 24-30 26352872-0 2015 ABCC5 and ABCG1 polymorphisms predict irinotecan-induced severe toxicity in metastatic colorectal cancer patients. Irinotecan 38-48 ATP binding cassette subfamily C member 5 Homo sapiens 0-5 26352872-0 2015 ABCC5 and ABCG1 polymorphisms predict irinotecan-induced severe toxicity in metastatic colorectal cancer patients. Irinotecan 38-48 ATP binding cassette subfamily G member 1 Homo sapiens 10-15 26352872-4 2015 MATERIALS AND METHODS: In this study, we aimed to discover toxicity-associated markers in seven transporter genes participating in irinotecan pharmacokinetics involving the ABC transporter genes ABCB1, ABCC1, ABCC2, ABCC5, ABCG1, and ABCG2 and the solute carrier organic anion transporter gene SLCO1B1 and using a haplotype-tagging single-nucleotide polymorphisms (n=210 htSNPs) strategy. Irinotecan 131-141 ATP binding cassette subfamily B member 1 Homo sapiens 195-200 26352872-4 2015 MATERIALS AND METHODS: In this study, we aimed to discover toxicity-associated markers in seven transporter genes participating in irinotecan pharmacokinetics involving the ABC transporter genes ABCB1, ABCC1, ABCC2, ABCC5, ABCG1, and ABCG2 and the solute carrier organic anion transporter gene SLCO1B1 and using a haplotype-tagging single-nucleotide polymorphisms (n=210 htSNPs) strategy. Irinotecan 131-141 ATP binding cassette subfamily C member 1 Homo sapiens 202-207 26352872-4 2015 MATERIALS AND METHODS: In this study, we aimed to discover toxicity-associated markers in seven transporter genes participating in irinotecan pharmacokinetics involving the ABC transporter genes ABCB1, ABCC1, ABCC2, ABCC5, ABCG1, and ABCG2 and the solute carrier organic anion transporter gene SLCO1B1 and using a haplotype-tagging single-nucleotide polymorphisms (n=210 htSNPs) strategy. Irinotecan 131-141 ATP binding cassette subfamily C member 2 Homo sapiens 209-214 26352872-4 2015 MATERIALS AND METHODS: In this study, we aimed to discover toxicity-associated markers in seven transporter genes participating in irinotecan pharmacokinetics involving the ABC transporter genes ABCB1, ABCC1, ABCC2, ABCC5, ABCG1, and ABCG2 and the solute carrier organic anion transporter gene SLCO1B1 and using a haplotype-tagging single-nucleotide polymorphisms (n=210 htSNPs) strategy. Irinotecan 131-141 ATP binding cassette subfamily C member 5 Homo sapiens 216-221 26352872-4 2015 MATERIALS AND METHODS: In this study, we aimed to discover toxicity-associated markers in seven transporter genes participating in irinotecan pharmacokinetics involving the ABC transporter genes ABCB1, ABCC1, ABCC2, ABCC5, ABCG1, and ABCG2 and the solute carrier organic anion transporter gene SLCO1B1 and using a haplotype-tagging single-nucleotide polymorphisms (n=210 htSNPs) strategy. Irinotecan 131-141 ATP binding cassette subfamily G member 1 Homo sapiens 223-228 26352872-4 2015 MATERIALS AND METHODS: In this study, we aimed to discover toxicity-associated markers in seven transporter genes participating in irinotecan pharmacokinetics involving the ABC transporter genes ABCB1, ABCC1, ABCC2, ABCC5, ABCG1, and ABCG2 and the solute carrier organic anion transporter gene SLCO1B1 and using a haplotype-tagging single-nucleotide polymorphisms (n=210 htSNPs) strategy. Irinotecan 131-141 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 234-239 26352872-4 2015 MATERIALS AND METHODS: In this study, we aimed to discover toxicity-associated markers in seven transporter genes participating in irinotecan pharmacokinetics involving the ABC transporter genes ABCB1, ABCC1, ABCC2, ABCC5, ABCG1, and ABCG2 and the solute carrier organic anion transporter gene SLCO1B1 and using a haplotype-tagging single-nucleotide polymorphisms (n=210 htSNPs) strategy. Irinotecan 131-141 solute carrier organic anion transporter family member 1B1 Homo sapiens 294-301 26431797-0 2015 CPT-11 activates NLRP3 inflammasome through JNK and NF-kappaB signalings. Irinotecan 0-6 NLR family pyrin domain containing 3 Homo sapiens 17-22 26431797-0 2015 CPT-11 activates NLRP3 inflammasome through JNK and NF-kappaB signalings. Irinotecan 0-6 mitogen-activated protein kinase 8 Homo sapiens 44-47 26431797-0 2015 CPT-11 activates NLRP3 inflammasome through JNK and NF-kappaB signalings. Irinotecan 0-6 nuclear factor kappa B subunit 1 Homo sapiens 52-61 26431797-5 2015 In this study, we have discovered that CPT-11 promotes macrophage infiltration into intestinal tissues and activates the NOD-like receptor family, pyrin domain containing 3 (NLRP3) inflammasome, resulting in a robust IL-1beta response and colonic inflammation similar to DSS (dextran sodium sulfate) induced experimental colitis. Irinotecan 39-45 NLR family pyrin domain containing 3 Homo sapiens 174-179 26431797-5 2015 In this study, we have discovered that CPT-11 promotes macrophage infiltration into intestinal tissues and activates the NOD-like receptor family, pyrin domain containing 3 (NLRP3) inflammasome, resulting in a robust IL-1beta response and colonic inflammation similar to DSS (dextran sodium sulfate) induced experimental colitis. Irinotecan 39-45 interleukin 1 beta Homo sapiens 217-225 25778466-0 2015 A novel UGT1 marker associated with better tolerance against irinotecan-induced severe neutropenia in metastatic colorectal cancer patients. Irinotecan 61-71 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 8-12 25778466-1 2015 The risk of severe irinotecan-induced neutropenia has been shown to be related to the UGT1 variant UGT1A1*28, which increases exposure to the potent metabolite SN-38. Irinotecan 19-29 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 86-90 25778466-1 2015 The risk of severe irinotecan-induced neutropenia has been shown to be related to the UGT1 variant UGT1A1*28, which increases exposure to the potent metabolite SN-38. Irinotecan 19-29 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 99-105 25778466-1 2015 The risk of severe irinotecan-induced neutropenia has been shown to be related to the UGT1 variant UGT1A1*28, which increases exposure to the potent metabolite SN-38. Irinotecan 160-165 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 86-90 25778466-1 2015 The risk of severe irinotecan-induced neutropenia has been shown to be related to the UGT1 variant UGT1A1*28, which increases exposure to the potent metabolite SN-38. Irinotecan 160-165 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 99-105 26692528-10 2015 CONCLUSIONS: Our study showed that mCRC patients with UGT1A1 *1/*1 and *1/*28 genotypes could receive escalated doses of irinotecan to obtain a more favorable clinical outcome without significant AEs. Irinotecan 121-131 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 54-60 26692528-1 2015 PURPOSE: This study aimed to identify the efficacy and toxicity of the FOLFIRI regimen (fluorouracil, leucovorin, and irinotecan) with irinotecan dose escalation plus bevacizumab as first-line chemotherapy for metastatic colorectal cancer (mCRC) via UGT1A1 genotyping. Irinotecan 135-145 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 250-256 26552750-7 2015 Orthotopic HT-29 xenografts treated with standard CRC chemotherapeutics 5-fluorouracil, irinotecan, or oxaliplatin showed dramatic increases in CD26 compared to untreated tumors. Irinotecan 88-98 dipeptidyl peptidase 4 Homo sapiens 144-148 26142736-3 2015 As a target of 5-FU, thymidylate synthase (TS) expression level might be influenced by irinotecan. Irinotecan 87-97 thymidylate synthetase Homo sapiens 21-41 26142736-3 2015 As a target of 5-FU, thymidylate synthase (TS) expression level might be influenced by irinotecan. Irinotecan 87-97 thymidylate synthetase Homo sapiens 43-45 26142736-4 2015 Understanding whether this influence of TS is related with MMR status is helpful to the further exploration of the mechanism of irinotecan sensitivity in metastatic colon cancer with different MMR status. Irinotecan 128-138 thymidylate synthetase Homo sapiens 40-42 26142736-9 2015 Regulation of TS by irinotecan was evaluated by western blotting and quantitative real-time PCR assay. Irinotecan 20-30 thymidylate synthetase Homo sapiens 14-16 26142736-12 2015 TS expression level was reduced more in dMMR cells after irinotecan treatment (p < 0.05). Irinotecan 57-67 thymidylate synthetase Homo sapiens 0-2 26452027-10 2015 Another patient had a newly emerged PIK3CA H1047R mutation on cfDNA analysis at progression during cetuximab/irinotecan chemotherapy with gradual increase in allele frequency from 0.8 to 2.1%. Irinotecan 109-119 phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha Homo sapiens 36-42 26599335-7 2015 Enhanced effects were observed when regorafenib was combined with irradiation and irinotecan against PDGFRA amplified IGRG93 glioma and IGRM57 medulloblastoma respectively, resulting in 100% tumor regressions. Irinotecan 82-92 platelet derived growth factor receptor alpha Homo sapiens 101-107 26504021-1 2015 BACKGROUND: The combination of irinotecan, a topoisomerase I inhibitor with cetuximab, an antibody against epidermal growth factor receptor, produces synergistic and beneficial effects in patients with irinotecan-refractory colorectal cancer. Irinotecan 31-41 epidermal growth factor receptor Homo sapiens 107-139 26461062-10 2015 In our exploratory analysis anti-angiogenic/oxaliplatin-based regimens and anti-EGFR/irinotecan-based regimens were associated with the highest pR. Irinotecan 85-95 epidermal growth factor receptor Homo sapiens 80-84 26529243-0 2015 Human Lipocalin-Type Prostaglandin D Synthase-Based Drug Delivery System for Poorly Water-Soluble Anti-Cancer Drug SN-38. Irinotecan 115-120 prostaglandin D2 synthase Homo sapiens 6-45 26529243-3 2015 In this study, we show the development of a drug delivery system using L-PGDS, one that enables the direct clinical use of 7-ethyl-10-hydroxy-camptothecin (SN-38), a poorly water-soluble anti-cancer drug. Irinotecan 156-161 prostaglandin D2 synthase Homo sapiens 71-77 26529243-4 2015 In the presence of 2 mM L-PGDS, the concentration of SN-38 in PBS increased 1,130-fold as compared with that in PBS. Irinotecan 53-58 prostaglandin D2 synthase Homo sapiens 24-30 26529243-5 2015 Calorimetric experiments revealed that L-PGDS bound SN-38 at a molecular ratio of 1:3 with a dissociation constant value of 60 muM. Irinotecan 52-57 prostaglandin D2 synthase Homo sapiens 39-45 26529243-6 2015 The results of an in vitro growth inhibition assay revealed that the SN-38/L-PGDS complexes showed high anti-tumor activity against 3 human cancer cell lines, i.e., Colo201, MDA-MB-231, and PC-3 with a potency similar to that of SN-38 used alone. Irinotecan 69-74 prostaglandin D2 synthase Homo sapiens 75-81 26529243-6 2015 The results of an in vitro growth inhibition assay revealed that the SN-38/L-PGDS complexes showed high anti-tumor activity against 3 human cancer cell lines, i.e., Colo201, MDA-MB-231, and PC-3 with a potency similar to that of SN-38 used alone. Irinotecan 229-234 prostaglandin D2 synthase Homo sapiens 75-81 26529243-7 2015 The intravenous administration of SN-38/L-PGDS complexes to mice bearing Colo201 tumors showed a pronounced anti-tumor effect. Irinotecan 34-39 prostaglandin D2 synthase (brain) Mus musculus 40-46 26529243-9 2015 Taken together, L-PGDS enables the direct usage of SN-38 with reduced side effects. Irinotecan 51-56 prostaglandin D2 synthase Homo sapiens 16-22 26504021-1 2015 BACKGROUND: The combination of irinotecan, a topoisomerase I inhibitor with cetuximab, an antibody against epidermal growth factor receptor, produces synergistic and beneficial effects in patients with irinotecan-refractory colorectal cancer. Irinotecan 202-212 epidermal growth factor receptor Homo sapiens 107-139 26504021-9 2015 CONCLUSION: SN-38/cetuximab combination has synergistic anti-angiogenesis and anti-invasion activities mediated by down-regulation of phosphatidylinositol-3-kinases/AKT and mitogen-activated protein kinase/ERK pathways. Irinotecan 12-17 AKT serine/threonine kinase 1 Homo sapiens 165-168 26504021-9 2015 CONCLUSION: SN-38/cetuximab combination has synergistic anti-angiogenesis and anti-invasion activities mediated by down-regulation of phosphatidylinositol-3-kinases/AKT and mitogen-activated protein kinase/ERK pathways. Irinotecan 12-17 mitogen-activated protein kinase 3 Homo sapiens 206-209 26692923-6 2015 Lastly, in contrast to both SN-38 (active metabolite of irinotecan) and topotecan are substrates of the efflux pump proteins P-gp/MDR1 and ABCG2/BCRP, FL118 is not a substrate of P-gp and ABCG2. Irinotecan 28-33 phosphoglycolate phosphatase Homo sapiens 125-129 26271675-0 2015 HSP90 Inhibitor-SN-38 Conjugate Strategy for Targeted Delivery of Topoisomerase I Inhibitor to Tumors. Irinotecan 16-21 heat shock protein 90 alpha family class A member 1 Homo sapiens 0-5 26271675-6 2015 In vivo modeling showed that the HSP90 inhibitor moiety was required for selective retention of STA-12-8666, and this enrichment promoted extended release of active SN-38 within the tumor compartment. Irinotecan 165-170 heat shock protein 90 alpha family class A member 1 Homo sapiens 33-38 26216446-0 2015 Restoration of CBX7 expression increases the susceptibility of human lung carcinoma cells to irinotecan treatment. Irinotecan 93-103 chromobox 7 Homo sapiens 15-19 26216446-6 2015 Taken together, these results suggest that the retention of CBX7 expression may play a role in the modulation of chemosensitivity of lung cancer patients to the treatment with irinotecan and etoposide. Irinotecan 176-186 chromobox 7 Homo sapiens 60-64 26429709-7 2015 The study was amended to exclude 4 UGT1A1*28 7/7 homozygous patients due to frequent irinotecan-related grade 3/grade 4 diarrhea and/or febrile neutropenia. Irinotecan 85-95 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 35-41 26509550-0 2015 Effect of Cellular Location of Human Carboxylesterase 2 on CPT-11 Hydrolysis and Anticancer Activity. Irinotecan 59-65 carboxylesterase 2 Homo sapiens 37-55 26509550-2 2015 Hydrolysis of CPT-11 by human carboxylesterase 2 (CE2) generates SN-38, a topoisomerase I inhibitor that is the active anti-tumor agent. Irinotecan 14-20 carboxylesterase 2 Homo sapiens 30-48 26509550-2 2015 Hydrolysis of CPT-11 by human carboxylesterase 2 (CE2) generates SN-38, a topoisomerase I inhibitor that is the active anti-tumor agent. Irinotecan 14-20 carboxylesterase 2 Homo sapiens 50-53 26509550-2 2015 Hydrolysis of CPT-11 by human carboxylesterase 2 (CE2) generates SN-38, a topoisomerase I inhibitor that is the active anti-tumor agent. Irinotecan 65-70 carboxylesterase 2 Homo sapiens 30-48 26509550-2 2015 Hydrolysis of CPT-11 by human carboxylesterase 2 (CE2) generates SN-38, a topoisomerase I inhibitor that is the active anti-tumor agent. Irinotecan 65-70 carboxylesterase 2 Homo sapiens 50-53 26509550-9 2015 Cancer cells that expressed all three forms of CE2 were more sensitive to CPT-11 as compared to unmodified cancer cells, but the membrane-anchored and ER-retained forms of CE2 were consistently more effective than secreted CE2. Irinotecan 74-80 carboxylesterase 2 Homo sapiens 47-50 26509550-10 2015 We conclude that expression of CE2 in the ER or on the membrane of cancer cells is suitable for enhancing CPT-11 anticancer activity. Irinotecan 106-112 carboxylesterase 2 Homo sapiens 31-34 26692923-6 2015 Lastly, in contrast to both SN-38 (active metabolite of irinotecan) and topotecan are substrates of the efflux pump proteins P-gp/MDR1 and ABCG2/BCRP, FL118 is not a substrate of P-gp and ABCG2. Irinotecan 28-33 ATP binding cassette subfamily B member 1 Homo sapiens 130-134 26692923-6 2015 Lastly, in contrast to both SN-38 (active metabolite of irinotecan) and topotecan are substrates of the efflux pump proteins P-gp/MDR1 and ABCG2/BCRP, FL118 is not a substrate of P-gp and ABCG2. Irinotecan 28-33 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 139-144 26692923-6 2015 Lastly, in contrast to both SN-38 (active metabolite of irinotecan) and topotecan are substrates of the efflux pump proteins P-gp/MDR1 and ABCG2/BCRP, FL118 is not a substrate of P-gp and ABCG2. Irinotecan 28-33 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 145-149 26692923-6 2015 Lastly, in contrast to both SN-38 (active metabolite of irinotecan) and topotecan are substrates of the efflux pump proteins P-gp/MDR1 and ABCG2/BCRP, FL118 is not a substrate of P-gp and ABCG2. Irinotecan 56-66 phosphoglycolate phosphatase Homo sapiens 125-129 26692923-6 2015 Lastly, in contrast to both SN-38 (active metabolite of irinotecan) and topotecan are substrates of the efflux pump proteins P-gp/MDR1 and ABCG2/BCRP, FL118 is not a substrate of P-gp and ABCG2. Irinotecan 56-66 ATP binding cassette subfamily B member 1 Homo sapiens 130-134 26692923-6 2015 Lastly, in contrast to both SN-38 (active metabolite of irinotecan) and topotecan are substrates of the efflux pump proteins P-gp/MDR1 and ABCG2/BCRP, FL118 is not a substrate of P-gp and ABCG2. Irinotecan 56-66 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 139-144 26315125-0 2015 Protein corona hampers targeting potential of MUC1 aptamer functionalized SN-38 core-shell nanoparticles. Irinotecan 74-79 mucin 1, cell surface associated Homo sapiens 46-50 26315125-3 2015 SN-38, an active metabolite of camptothecin, conjugated to hyaluronic acid (HA) was used as the shell of chitosan nanoparticles decorated with MUC1 aptamer. Irinotecan 0-5 mucin 1, cell surface associated Homo sapiens 143-147 26463521-3 2015 Conventional irinotecan distributes poorly to brain metastases; therefore, NKTR-102, a PEGylated irinotecan conjugate should enhance irinotecan and its active metabolite SN38 exposure in brain metastases leading to brain tumor cytotoxicity. Irinotecan 97-107 natural killer tumor recognition sequence Mus musculus 75-79 26334566-4 2015 SN-38G formation by Ugt1a1 enzyme was analyzed in liver S9 fractions. Irinotecan 0-5 UDP glucuronosyltransferase 1 family, polypeptide A1 Mus musculus 20-26 26334566-10 2015 Conversion of SN-38 to SN-38G by Ugt1a1 enzyme was reduced by ~2-fold in liver S9 fractions in DIO mice. Irinotecan 14-19 UDP glucuronosyltransferase 1 family, polypeptide A1 Mus musculus 33-39 26334566-10 2015 Conversion of SN-38 to SN-38G by Ugt1a1 enzyme was reduced by ~2-fold in liver S9 fractions in DIO mice. Irinotecan 23-28 UDP glucuronosyltransferase 1 family, polypeptide A1 Mus musculus 33-39 26334566-13 2015 SIGNIFICANCE: Our study demonstrates that reduced hepatic Ugt1a activity during obesity likely contributes to reduced glucuronidation, and results in higher levels of the toxic metabolite, SN-38. Irinotecan 189-194 Ugt1a@ Mus musculus 58-63 26600773-2 2015 Bevacizumab, a monoclonal antibody to VEGF, in common use as an adjunct to standard chemotherapy like irinotecan in advanced colorectal cancer, also affects the normal (nontumor) vasculature. Irinotecan 102-112 vascular endothelial growth factor A Homo sapiens 38-42 26463521-3 2015 Conventional irinotecan distributes poorly to brain metastases; therefore, NKTR-102, a PEGylated irinotecan conjugate should enhance irinotecan and its active metabolite SN38 exposure in brain metastases leading to brain tumor cytotoxicity. Irinotecan 97-107 natural killer tumor recognition sequence Mus musculus 75-79 26440613-12 2015 This study shows a critical role of the MyD88-mediated TLR2 and TLR9 signaling in the pathogenesis of irinotecan-induced intestinal mucositis. Irinotecan 102-112 toll-like receptor 9 Mus musculus 64-68 25891902-3 2015 RS1 is in the catalytic loop, RS2 is the Phe in the DFG motif, RS3 is at the C-terminus of the alphaC-Helix, and RS4 is at the beginning of beta4. Irinotecan 113-116 tubulin beta 3 class III Homo sapiens 140-145 26440613-0 2015 The Adaptor Protein Myd88 Is a Key Signaling Molecule in the Pathogenesis of Irinotecan-Induced Intestinal Mucositis. Irinotecan 77-87 myeloid differentiation primary response gene 88 Mus musculus 20-25 26440613-4 2015 Then, we evaluated the involvement of TLRs and MyD88 in the pathogenesis of irinotecan-induced intestinal mucositis. Irinotecan 76-86 myeloid differentiation primary response gene 88 Mus musculus 47-52 26440613-9 2015 Genetic deletion of MyD88, TLR2 or TLR9 effectively controlled the signs of intestinal injury when compared with irinotecan-administered WT controls (P<0.05). Irinotecan 113-123 myeloid differentiation primary response gene 88 Mus musculus 20-25 26440613-10 2015 In contrast to the MyD88-/- and TLR2-/- mice, the irinotecan-injected TLR9-/- mice showed a reduced survival, a marked diarrhea and an enhanced expression of IL-18 versus irinotecan-injected WT controls. Irinotecan 50-60 toll-like receptor 9 Mus musculus 70-74 26440613-10 2015 In contrast to the MyD88-/- and TLR2-/- mice, the irinotecan-injected TLR9-/- mice showed a reduced survival, a marked diarrhea and an enhanced expression of IL-18 versus irinotecan-injected WT controls. Irinotecan 50-60 interleukin 18 Mus musculus 158-163 26440613-10 2015 In contrast to the MyD88-/- and TLR2-/- mice, the irinotecan-injected TLR9-/- mice showed a reduced survival, a marked diarrhea and an enhanced expression of IL-18 versus irinotecan-injected WT controls. Irinotecan 171-181 toll-like receptor 9 Mus musculus 70-74 26440613-12 2015 This study shows a critical role of the MyD88-mediated TLR2 and TLR9 signaling in the pathogenesis of irinotecan-induced intestinal mucositis. Irinotecan 102-112 myeloid differentiation primary response gene 88 Mus musculus 40-45 26440613-12 2015 This study shows a critical role of the MyD88-mediated TLR2 and TLR9 signaling in the pathogenesis of irinotecan-induced intestinal mucositis. Irinotecan 102-112 toll-like receptor 2 Mus musculus 55-59 26196680-8 2015 SW480 CRC cells that naturally express Lgr5 are those that are floating, and they are more sensitive to the chemotherapeutic compounds irinotecan (maximum difference approximately two times, 0.0001<P<0.0052) and oxaliplatin (maximum difference ~1.5 times, 0.0001<P<0.0024) than Lgr5-negative (attached) SW480 cells. Irinotecan 135-145 leucine rich repeat containing G protein-coupled receptor 5 Homo sapiens 39-43 26196680-8 2015 SW480 CRC cells that naturally express Lgr5 are those that are floating, and they are more sensitive to the chemotherapeutic compounds irinotecan (maximum difference approximately two times, 0.0001<P<0.0052) and oxaliplatin (maximum difference ~1.5 times, 0.0001<P<0.0024) than Lgr5-negative (attached) SW480 cells. Irinotecan 135-145 leucine rich repeat containing G protein-coupled receptor 5 Homo sapiens 290-294 26196680-11 2015 Transfected Lgr5-overexpressing attached cells showed similar results with irinotecan and oxaliplatin, confirming that the detected differences in sensitivity to these drugs were Lgr5 driven. Irinotecan 75-85 leucine rich repeat containing G protein-coupled receptor 5 Homo sapiens 12-16 24724613-0 2015 Interleukin-18 as a target for modulation of irinotecan-induced intestinal toxicity: a step towards a better therapeutic index? Irinotecan 45-55 interleukin 18 Homo sapiens 0-14 26316041-6 2015 Knockdown of E-cadherin by lentiviral delivery of shRNA significantly reduced chemosensitivity to 5-fluorouracil and irinotecan, increased beta-catenin protein level in HCT116 3D cultures. Irinotecan 117-127 cadherin 1 Homo sapiens 13-23 26090722-7 2015 These results suggest that HGF produced by fibroblast induce CPT-11 resistance, and that anti-HGF antibody abrogate such resistance in vivo. Irinotecan 61-67 hepatocyte growth factor Homo sapiens 27-30 26316041-8 2015 Knockdown of beta-catenin significantly increased chemosensitivity to 5-fluorouracil and irinotecan in HCT116 3D cultures and LoVo 3D cultures. Irinotecan 89-99 catenin beta 1 Homo sapiens 13-25 26224606-2 2015 Therefore, the aim of our study was to evaluate the differences in phenotype between germ-free (GF) and conventional (CV) mice, and the role of beta-glucuronidase-producing bacteria in the development of irinotecan treatment in a murine model. Irinotecan 204-214 glucuronidase, beta Mus musculus 144-162 26486455-0 2015 Exploratory biomarker analysis for treatment response in KRAS wild type metastatic colorectal cancer patients who received cetuximab plus irinotecan. Irinotecan 138-148 KRAS proto-oncogene, GTPase Homo sapiens 57-61 26090722-0 2015 Humanized anti-hepatocyte growth factor (HGF) antibody suppresses innate irinotecan (CPT-11) resistance induced by fibroblast-derived HGF. Irinotecan 73-83 hepatocyte growth factor Homo sapiens 10-39 26090722-0 2015 Humanized anti-hepatocyte growth factor (HGF) antibody suppresses innate irinotecan (CPT-11) resistance induced by fibroblast-derived HGF. Irinotecan 73-83 hepatocyte growth factor Homo sapiens 41-44 26090722-0 2015 Humanized anti-hepatocyte growth factor (HGF) antibody suppresses innate irinotecan (CPT-11) resistance induced by fibroblast-derived HGF. Irinotecan 73-83 hepatocyte growth factor Homo sapiens 134-137 26090722-0 2015 Humanized anti-hepatocyte growth factor (HGF) antibody suppresses innate irinotecan (CPT-11) resistance induced by fibroblast-derived HGF. Irinotecan 85-91 hepatocyte growth factor Homo sapiens 10-39 26090722-0 2015 Humanized anti-hepatocyte growth factor (HGF) antibody suppresses innate irinotecan (CPT-11) resistance induced by fibroblast-derived HGF. Irinotecan 85-91 hepatocyte growth factor Homo sapiens 41-44 26090722-0 2015 Humanized anti-hepatocyte growth factor (HGF) antibody suppresses innate irinotecan (CPT-11) resistance induced by fibroblast-derived HGF. Irinotecan 85-91 hepatocyte growth factor Homo sapiens 134-137 26090722-4 2015 Combination treatment of anti-HGF antibody and irinotecan (CPT-11) directly enhanced CPT-11 sensitivity in CRC. Irinotecan 85-91 hepatocyte growth factor Homo sapiens 30-33 26206183-6 2015 Under hypoxic growth condition, ABCG2 and ABCB1-overexpressing cells sorted out by FACS technique as side population (SP) were found to be significantly more responsive to SN-38 (ABCG2 and ABCB1 substrate anticancer drug) in the presence of vatalanib. Irinotecan 172-177 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 32-37 26109630-2 2015 Patients with no response to first-line treatment with S-1 often receive a taxane-alone or irinotecan-alone as second-line treatment. Irinotecan 91-101 proteasome 26S subunit, non-ATPase 1 Homo sapiens 55-58 26109630-3 2015 However, second-line treatment with S-1 plus irinotecan is widely used in patients with AGC resistant to first-line S-1-based chemotherapy. Irinotecan 45-55 proteasome 26S subunit, non-ATPase 1 Homo sapiens 116-119 26206183-6 2015 Under hypoxic growth condition, ABCG2 and ABCB1-overexpressing cells sorted out by FACS technique as side population (SP) were found to be significantly more responsive to SN-38 (ABCG2 and ABCB1 substrate anticancer drug) in the presence of vatalanib. Irinotecan 172-177 ATP binding cassette subfamily B member 1 Homo sapiens 42-47 26101915-2 2015 The rational development of IMMU-132 represents a paradigm shift as an ADC that binds a well-known moderately-cytotoxic drug, SN-38, to the anti-Trop-2 antibody. Irinotecan 126-131 tumor associated calcium signal transducer 2 Homo sapiens 145-151 26516354-1 2015 BACKGROUND: Active efflux of irinotecan by ATP-binding cassette (ABC)-transporters, in particular ABCB1 and ABCG2, is a well-established drug resistance mechanism in vitro and in pre-clinical mouse models, but its relevance in colorectal cancer (CRC) patients is unknown. Irinotecan 29-39 ATP-binding cassette, sub-family B (MDR/TAP), member 1B Mus musculus 98-103 26516354-1 2015 BACKGROUND: Active efflux of irinotecan by ATP-binding cassette (ABC)-transporters, in particular ABCB1 and ABCG2, is a well-established drug resistance mechanism in vitro and in pre-clinical mouse models, but its relevance in colorectal cancer (CRC) patients is unknown. Irinotecan 29-39 ATP binding cassette subfamily G member 2 (Junior blood group) Mus musculus 108-113 26516354-2 2015 Therefore, we assessed the association between ABC-transporter expression and tumour response to irinotecan in patients with metastatic CRC. Irinotecan 97-107 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 47-62 26516354-4 2015 Kaplan-Meier and Cox proportional hazard regression analyses were performed to assess the association of ABC transporter expression with irinotecan response. Irinotecan 137-147 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 105-120 26206183-6 2015 Under hypoxic growth condition, ABCG2 and ABCB1-overexpressing cells sorted out by FACS technique as side population (SP) were found to be significantly more responsive to SN-38 (ABCG2 and ABCB1 substrate anticancer drug) in the presence of vatalanib. Irinotecan 172-177 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 179-184 26206183-6 2015 Under hypoxic growth condition, ABCG2 and ABCB1-overexpressing cells sorted out by FACS technique as side population (SP) were found to be significantly more responsive to SN-38 (ABCG2 and ABCB1 substrate anticancer drug) in the presence of vatalanib. Irinotecan 172-177 ATP binding cassette subfamily B member 1 Homo sapiens 189-194 25934339-1 2015 The purpose of this study is to evaluate the influence of germline polymorphisms of cytochrome P450 (CYP450) on objective response, progression-free survival (PFS) and overall suruvival (OS) in metastatic colorectal cancer (mCRC) receiving the combination chemotherapy of irinotecan, 5-fluorouracil, and leucovorin (FOLFIRI). Irinotecan 272-282 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-99 25782327-0 2015 DPD and UGT1A1 deficiency in colorectal cancer patients receiving triplet chemotherapy with fluoropyrimidines, oxaliplatin and irinotecan. Irinotecan 127-137 dihydropyrimidine dehydrogenase Homo sapiens 0-3 25782327-0 2015 DPD and UGT1A1 deficiency in colorectal cancer patients receiving triplet chemotherapy with fluoropyrimidines, oxaliplatin and irinotecan. Irinotecan 127-137 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 8-14 25782327-2 2015 Single nucleotide polymorphisms (SNPs) in DPYD and UGT1A1 influence fluoropyrimdines and irinotecan adverse events (AEs). Irinotecan 89-99 dihydropyrimidine dehydrogenase Homo sapiens 42-46 25782327-2 2015 Single nucleotide polymorphisms (SNPs) in DPYD and UGT1A1 influence fluoropyrimdines and irinotecan adverse events (AEs). Irinotecan 89-99 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 51-57 26141947-8 2015 Irinotecan cytotoxicity was directly linked to clock gene BMAL1 expression: The least apoptosis resulted from drug exposure near BMAL1 mRNA nadir (P < 0.001), whereas clock silencing through siBMAL1 exposure ablated all the chronopharmacology mechanisms. Irinotecan 0-10 aryl hydrocarbon receptor nuclear translocator like Homo sapiens 58-63 26141947-8 2015 Irinotecan cytotoxicity was directly linked to clock gene BMAL1 expression: The least apoptosis resulted from drug exposure near BMAL1 mRNA nadir (P < 0.001), whereas clock silencing through siBMAL1 exposure ablated all the chronopharmacology mechanisms. Irinotecan 0-10 aryl hydrocarbon receptor nuclear translocator like Homo sapiens 129-134 25944802-0 2015 First-in-Human Trial of a Novel Anti-Trop-2 Antibody-SN-38 Conjugate, Sacituzumab Govitecan, for the Treatment of Diverse Metastatic Solid Tumors. Irinotecan 53-58 tumor associated calcium signal transducer 2 Homo sapiens 37-43 25944802-1 2015 PURPOSE: Sacituzumab govitecan (IMMU-132) is an antibody-drug conjugate (ADC) targeting Trop-2, a surface glycoprotein expressed on many epithelial tumors, for delivery of SN-38, the active metabolite of irinotecan. Irinotecan 172-177 tumor associated calcium signal transducer 2 Homo sapiens 88-94 25944802-1 2015 PURPOSE: Sacituzumab govitecan (IMMU-132) is an antibody-drug conjugate (ADC) targeting Trop-2, a surface glycoprotein expressed on many epithelial tumors, for delivery of SN-38, the active metabolite of irinotecan. Irinotecan 204-214 tumor associated calcium signal transducer 2 Homo sapiens 88-94 26713521-2 2015 METHODS: SRB colorimetry was employed to measure the viability of HepG-2 and BEL-7402 cells after the treatment of SN-38 with sorafenib. Irinotecan 115-120 chaperonin containing TCP1 subunit 4 Homo sapiens 9-12 25934339-1 2015 The purpose of this study is to evaluate the influence of germline polymorphisms of cytochrome P450 (CYP450) on objective response, progression-free survival (PFS) and overall suruvival (OS) in metastatic colorectal cancer (mCRC) receiving the combination chemotherapy of irinotecan, 5-fluorouracil, and leucovorin (FOLFIRI). Irinotecan 272-282 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-107 26713521-5 2015 RESULTS: SRB colorimetry showed the synergistic anticancer activities of SN-38 combined with sorafenib, with a combination index of <0.9. Irinotecan 73-78 chaperonin containing TCP1 subunit 4 Homo sapiens 9-12 26229432-0 2015 Relationship between UGT1A1*6/*28 polymorphisms and severe toxicities in Chinese patients with pancreatic or biliary tract cancer treated with irinotecan-containing regimens. Irinotecan 143-153 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 21-27 26275292-16 2015 CONCLUSION AND RELEVANCE: The addition of EGFR-I to irinotecan-based chemotherapy has consistent efficacy, regardless of FP regimen, whereas EGFR-I and oxaliplatin-based regimens were most active with infusional 5FU. Irinotecan 52-62 epidermal growth factor receptor Homo sapiens 42-46 26257651-0 2015 Simultaneous inhibition of ATR and PARP sensitizes colon cancer cell lines to irinotecan. Irinotecan 78-88 ATR serine/threonine kinase Homo sapiens 27-30 26257651-0 2015 Simultaneous inhibition of ATR and PARP sensitizes colon cancer cell lines to irinotecan. Irinotecan 78-88 poly(ADP-ribose) polymerase 1 Homo sapiens 35-39 26229432-1 2015 PURPOSE: The aim of this retrospective study was to investigate the relationship between UGT1A1 polymorphisms and toxicities in Chinese patients with pancreatic or biliary tract cancer receiving irinotecan-containing regimens as the second- or third-line chemotherapy. Irinotecan 195-205 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 89-95 26229432-12 2015 CONCLUSION: In Chinese patients with pancreatic or biliary tract cancer administered irinotecan-containing regimens, those with UGT1A1*6 variant may have a high risk of severe neutropenia. Irinotecan 85-95 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 128-134 26125934-0 2015 Correlation of UGT1A1 and ERCC1 gene polymorphisms with the outcome of combined irinotecan plus cisplatin treatment in recurrent ovarian cancer. Irinotecan 80-90 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 15-21 25870101-5 2015 OTS167 glucuronidation in HLM was highly correlated with thyroxine glucuronidation (r = 0.91, P < 0.0001), SN-38 glucuronidation (r = 0.79, P < 0.0001), and UGT1A1 mRNA (r = 0.72, P < 0.0001). Irinotecan 110-115 oxysterol binding protein 2 Homo sapiens 26-29 25213123-0 2015 Involvement of matrix metalloproteinases (MMP-3 and MMP-9) in the pathogenesis of irinotecan-induced oral mucositis. Irinotecan 82-92 matrix metallopeptidase 3 Rattus norvegicus 42-47 25213123-0 2015 Involvement of matrix metalloproteinases (MMP-3 and MMP-9) in the pathogenesis of irinotecan-induced oral mucositis. Irinotecan 82-92 matrix metallopeptidase 9 Rattus norvegicus 52-57 25213123-3 2015 We investigated tissue expression of MMP-3 and MMP-9 over time in a pre-clinical model of irinotecan-induced oral mucositis (OM). Irinotecan 90-100 matrix metallopeptidase 3 Rattus norvegicus 37-42 25779651-3 2015 While irinotecan is known to stabilize the complex of topo I and DNA, the enzyme tyrosyl-DNA phosphodiesterase 1 (TDP-1) functions in an opposing manner by releasing topo I from DNA. Irinotecan 6-16 tyrosyl-DNA phosphodiesterase 1 Mus musculus 81-112 25779651-3 2015 While irinotecan is known to stabilize the complex of topo I and DNA, the enzyme tyrosyl-DNA phosphodiesterase 1 (TDP-1) functions in an opposing manner by releasing topo I from DNA. Irinotecan 6-16 tyrosyl-DNA phosphodiesterase 1 Mus musculus 114-119 24474168-5 2015 CPT-11 promoted the release of AIF from mitochondria and its translocation to the nucleus. Irinotecan 0-6 apoptosis inducing factor mitochondria associated 1 Homo sapiens 31-34 24474168-7 2015 Induction of cell-cycle arrest by CPT-11 was associated with changes in expression of key cell-cycle regulators such as CDK2, Chk2, and cyclin D in HL-60 cells. Irinotecan 34-40 cyclin dependent kinase 2 Homo sapiens 120-124 24474168-7 2015 Induction of cell-cycle arrest by CPT-11 was associated with changes in expression of key cell-cycle regulators such as CDK2, Chk2, and cyclin D in HL-60 cells. Irinotecan 34-40 checkpoint kinase 2 Homo sapiens 126-130 26591441-0 2015 [UGT1A1 Genotyping for Proper Use of Irinotecan]. Irinotecan 37-47 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 1-7 26591441-3 2015 Irinotecan is metabolized to form active SN-38, which is further conjugated and detoxified by the UDP-glucuronosyltransferase (UGT) 1A1 enzyme. Irinotecan 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 98-135 26591441-3 2015 Irinotecan is metabolized to form active SN-38, which is further conjugated and detoxified by the UDP-glucuronosyltransferase (UGT) 1A1 enzyme. Irinotecan 41-46 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 98-135 26591441-4 2015 Recent pharmacogenetic studies on irinotecan have revealed the impact of UGT1A1 polymorphisms on severe adverse effects. Irinotecan 34-44 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 73-79 26591441-5 2015 A variant in the promoter of the UGT1A1 gene, the UGT1A1 *28 allele, has been extensively studied, and pharmacogenetic relationships between the variant and severe toxicities of irinotecan have been reported. Irinotecan 178-188 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-39 26591441-5 2015 A variant in the promoter of the UGT1A1 gene, the UGT1A1 *28 allele, has been extensively studied, and pharmacogenetic relationships between the variant and severe toxicities of irinotecan have been reported. Irinotecan 178-188 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 50-56 26591441-8 2015 The concurrence of UGT1A1*28 and UGT1A1*6, even when heterozygous, markedly alters the disposition of irinotecan, potentially increasing toxicity, which is now written on the label of irinotecan in Japan. Irinotecan 102-112 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 19-25 26591441-8 2015 The concurrence of UGT1A1*28 and UGT1A1*6, even when heterozygous, markedly alters the disposition of irinotecan, potentially increasing toxicity, which is now written on the label of irinotecan in Japan. Irinotecan 102-112 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-39 26591441-8 2015 The concurrence of UGT1A1*28 and UGT1A1*6, even when heterozygous, markedly alters the disposition of irinotecan, potentially increasing toxicity, which is now written on the label of irinotecan in Japan. Irinotecan 184-194 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 19-25 26591441-8 2015 The concurrence of UGT1A1*28 and UGT1A1*6, even when heterozygous, markedly alters the disposition of irinotecan, potentially increasing toxicity, which is now written on the label of irinotecan in Japan. Irinotecan 184-194 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-39 26591441-9 2015 For patients showing homozygosity for UGT1A1*28, *6, or compound heterozygosity for UGT1A1*6 and *28, dose reduction of irinotecan is strongly recommended. Irinotecan 120-130 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 38-44 26591441-9 2015 For patients showing homozygosity for UGT1A1*28, *6, or compound heterozygosity for UGT1A1*6 and *28, dose reduction of irinotecan is strongly recommended. Irinotecan 120-130 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 84-90 26591441-12 2015 At present, irinotecan chemotherapy based on a patient"s UGT1A1 genetic status is scientifically reasonable. Irinotecan 12-22 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 57-63 26125934-0 2015 Correlation of UGT1A1 and ERCC1 gene polymorphisms with the outcome of combined irinotecan plus cisplatin treatment in recurrent ovarian cancer. Irinotecan 80-90 ERCC excision repair 1, endonuclease non-catalytic subunit Homo sapiens 26-31 26125934-1 2015 The aim of this study was to define the genotypes of UGT1A1 and ERCC1 and to examine their relationship with the efficacy and toxicity of a combination therapy of irinotecan and cisplatin in patients with advanced ovarian cancer. Irinotecan 163-173 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 53-59 26125934-1 2015 The aim of this study was to define the genotypes of UGT1A1 and ERCC1 and to examine their relationship with the efficacy and toxicity of a combination therapy of irinotecan and cisplatin in patients with advanced ovarian cancer. Irinotecan 163-173 ERCC excision repair 1, endonuclease non-catalytic subunit Homo sapiens 64-69 26125934-7 2015 Together, the results from this study suggest that UGT1A1 is a target gene for tardive diarrhea, and that the UGT1A1*28 gene mutation might increase the risk of diarrhea with irinotecan-based chemotherapy. Irinotecan 175-185 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 51-57 26125934-7 2015 Together, the results from this study suggest that UGT1A1 is a target gene for tardive diarrhea, and that the UGT1A1*28 gene mutation might increase the risk of diarrhea with irinotecan-based chemotherapy. Irinotecan 175-185 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 110-116 26125934-8 2015 Furthermore, the results suggest that ERCC1 WW carriers might obtain a better rate of clinical response from a combined irinotecan and cisplatin regimen than ERCC1 WM+MM carriers. Irinotecan 120-130 ERCC excision repair 1, endonuclease non-catalytic subunit Homo sapiens 38-43 25917289-4 2015 Our data indicated that both gefitinib and DMG are potent inhibitors of SN-38 glucuronidation via UGT1A1 inhibition. Irinotecan 72-77 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 98-104 25996449-3 2015 The nanoshells having surface environment modified by hexanethiol SAMs provided high capacity both for hydrophilic DNAzyme (Dz) to induce gene silencing and for hydrophobic SN38 (7-ethyl-10-hydroxycamptothecin), anticancer drug. Irinotecan 179-209 methionine adenosyltransferase 1A Homo sapiens 66-70 25777966-1 2015 INTRODUCTION: Topoisomerase 1 (TOP1) and 2A (TOP2A) are potential predictive biomarkers for irinotecan and anthracycline treatment, respectively, in colorectal cancer (CRC), and we have recently reported a high frequency of gene gain of the TOP1 and TOP2A genes in CRC. Irinotecan 92-102 DNA topoisomerase II alpha Homo sapiens 45-50 25817555-4 2015 The present review on the impact of the deficient UGT1A1*28 variant on irinotecan efficacy and toxicity was produced by a French joint workgroup comprising the Group of Clinical Onco-pharmacology (GPCO-Unicancer) and the National Pharmacogenetics Network (RNPGx). Irinotecan 71-81 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 50-56 25817555-5 2015 It clearly emerges that for irinotecan doses at least equal to 180 mg/m(2) , patients homozygous for the UGT1A1*28 allele are at increased risk of developing hematological and/or digestive toxicities. Irinotecan 28-38 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 105-111 25777966-1 2015 INTRODUCTION: Topoisomerase 1 (TOP1) and 2A (TOP2A) are potential predictive biomarkers for irinotecan and anthracycline treatment, respectively, in colorectal cancer (CRC), and we have recently reported a high frequency of gene gain of the TOP1 and TOP2A genes in CRC. Irinotecan 92-102 DNA topoisomerase II alpha Homo sapiens 250-255 25855895-10 2015 It was shown that they have a low cytotoxicity and the ability to reverse the BCRP-mediated SN-38 resistance. Irinotecan 92-97 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 78-82 25608838-0 2015 TIMP-1 and CEA as biomarkers in third-line treatment with irinotecan and cetuximab for metastatic colorectal cancer. Irinotecan 58-68 TIMP metallopeptidase inhibitor 1 Homo sapiens 0-6 25608838-0 2015 TIMP-1 and CEA as biomarkers in third-line treatment with irinotecan and cetuximab for metastatic colorectal cancer. Irinotecan 58-68 CEA cell adhesion molecule 3 Homo sapiens 11-14 25608838-5 2015 The aim of the present study was to investigate the clinical value of TIMP-1 in patients with metastatic colorectal cancer (mCRC) treated with cetuximab and irinotecan. Irinotecan 157-167 TIMP metallopeptidase inhibitor 1 Homo sapiens 70-76 26025324-0 2015 Carboxylesterase 2 as a Determinant of Response to Irinotecan and Neoadjuvant FOLFIRINOX Therapy in Pancreatic Ductal Adenocarcinoma. Irinotecan 51-61 carboxylesterase 2 Homo sapiens 0-18 26025324-2 2015 We have undertaken a comprehensive proteomic analysis to assess the differential expression and cellular localization of SHs, which uncovered distinctive expression of Carboxylesterase 2 (CES2), the most efficient carboxyl esterase in activating the prodrug irinotecan into SN-38, in pancreatic ductal adenocarcinoma (PDAC). Irinotecan 258-268 carboxylesterase 2 Homo sapiens 168-186 26025324-2 2015 We have undertaken a comprehensive proteomic analysis to assess the differential expression and cellular localization of SHs, which uncovered distinctive expression of Carboxylesterase 2 (CES2), the most efficient carboxyl esterase in activating the prodrug irinotecan into SN-38, in pancreatic ductal adenocarcinoma (PDAC). Irinotecan 258-268 carboxylesterase 2 Homo sapiens 188-192 26025324-2 2015 We have undertaken a comprehensive proteomic analysis to assess the differential expression and cellular localization of SHs, which uncovered distinctive expression of Carboxylesterase 2 (CES2), the most efficient carboxyl esterase in activating the prodrug irinotecan into SN-38, in pancreatic ductal adenocarcinoma (PDAC). Irinotecan 274-279 carboxylesterase 2 Homo sapiens 168-186 26025324-2 2015 We have undertaken a comprehensive proteomic analysis to assess the differential expression and cellular localization of SHs, which uncovered distinctive expression of Carboxylesterase 2 (CES2), the most efficient carboxyl esterase in activating the prodrug irinotecan into SN-38, in pancreatic ductal adenocarcinoma (PDAC). Irinotecan 274-279 carboxylesterase 2 Homo sapiens 188-192 26025324-5 2015 CES2 activity was assessed by monitoring the hydrolysis of the substrate p-NPA and correlated with irinotecan IC50 values by means of Pearson"s correlation. Irinotecan 99-109 carboxylesterase 2 Homo sapiens 0-4 26025324-9 2015 CES2 activity in 11 PDAC cell lines was inversely correlated with irinotecan IC50 values (R = -0.68, P = .02). Irinotecan 66-76 carboxylesterase 2 Homo sapiens 0-4 26025324-11 2015 CONCLUSION: Our findings suggest that CES2 expression and activity, by mediating the intratumoral activation of irinotecan, is a contributor to FOLFIRINOX sensitivity in pancreatic cancer and CES2 assessment may define a subset of patients likely to respond to irinotecan based therapy. Irinotecan 112-122 carboxylesterase 2 Homo sapiens 38-42 26025324-11 2015 CONCLUSION: Our findings suggest that CES2 expression and activity, by mediating the intratumoral activation of irinotecan, is a contributor to FOLFIRINOX sensitivity in pancreatic cancer and CES2 assessment may define a subset of patients likely to respond to irinotecan based therapy. Irinotecan 261-271 carboxylesterase 2 Homo sapiens 38-42 25880011-1 2015 BACKGROUND: In Asians, the risk of irinotecan-induced severe toxicities is related in part to UGT1A1*6 (UGT, UDP glucuronosyltransferase) and UGT1A1*28, variant alleles that reduce the elimination of SN-38, the active metabolite of irinotecan. Irinotecan 35-45 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 94-102 25915780-7 2015 Exposure of tumor cells to either free SN-38 or IMMU-132 demonstrated the same signaling pathways, with pJNK1/2 and p21(WAF1/Cip1) upregulation followed by cleavage of caspases 9, 7, and 3, ultimately leading to poly-ADP-ribose polymerase cleavage and double-stranded DNA breaks. Irinotecan 39-44 cyclin-dependent kinase inhibitor 1A (P21) Mus musculus 116-119 25915780-7 2015 Exposure of tumor cells to either free SN-38 or IMMU-132 demonstrated the same signaling pathways, with pJNK1/2 and p21(WAF1/Cip1) upregulation followed by cleavage of caspases 9, 7, and 3, ultimately leading to poly-ADP-ribose polymerase cleavage and double-stranded DNA breaks. Irinotecan 39-44 cyclin-dependent kinase inhibitor 1A (P21) Mus musculus 120-124 25915780-7 2015 Exposure of tumor cells to either free SN-38 or IMMU-132 demonstrated the same signaling pathways, with pJNK1/2 and p21(WAF1/Cip1) upregulation followed by cleavage of caspases 9, 7, and 3, ultimately leading to poly-ADP-ribose polymerase cleavage and double-stranded DNA breaks. Irinotecan 39-44 cyclin-dependent kinase inhibitor 1A (P21) Mus musculus 125-129 25915780-7 2015 Exposure of tumor cells to either free SN-38 or IMMU-132 demonstrated the same signaling pathways, with pJNK1/2 and p21(WAF1/Cip1) upregulation followed by cleavage of caspases 9, 7, and 3, ultimately leading to poly-ADP-ribose polymerase cleavage and double-stranded DNA breaks. Irinotecan 39-44 caspase 9 Mus musculus 168-188 25973791-0 2015 Identification of Sestrin3 Involved in the In vitro Resistance of Colorectal Cancer Cells to Irinotecan. Irinotecan 93-103 sestrin 3 Homo sapiens 18-26 25973791-5 2015 After gene ontology (GO) and KEGG pathway analysis of the microarray data, we specifically investigated whether Sestrin3 could decrease irinotecan resistance. Irinotecan 136-146 sestrin 3 Homo sapiens 112-120 25973791-6 2015 Our results revealed that Sestrin3 enhanced the anticancer effect of irinotecan in vitro in LoVo cells that had acquired resistance to irinotecan. Irinotecan 69-79 sestrin 3 Homo sapiens 26-34 25973791-6 2015 Our results revealed that Sestrin3 enhanced the anticancer effect of irinotecan in vitro in LoVo cells that had acquired resistance to irinotecan. Irinotecan 135-145 sestrin 3 Homo sapiens 26-34 25973791-8 2015 ROS production was increased by Sestrin3 knockdown in irinotecan-resistant LoVo cells. Irinotecan 54-64 sestrin 3 Homo sapiens 32-40 25973791-9 2015 Our results indicate that Sestrin3 might be a good target to develop therapeutics that can help to overcome resistance to irinotecan. Irinotecan 122-132 sestrin 3 Homo sapiens 26-34 25880011-1 2015 BACKGROUND: In Asians, the risk of irinotecan-induced severe toxicities is related in part to UGT1A1*6 (UGT, UDP glucuronosyltransferase) and UGT1A1*28, variant alleles that reduce the elimination of SN-38, the active metabolite of irinotecan. Irinotecan 35-45 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 94-97 25880011-1 2015 BACKGROUND: In Asians, the risk of irinotecan-induced severe toxicities is related in part to UGT1A1*6 (UGT, UDP glucuronosyltransferase) and UGT1A1*28, variant alleles that reduce the elimination of SN-38, the active metabolite of irinotecan. Irinotecan 35-45 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 94-100 25880011-1 2015 BACKGROUND: In Asians, the risk of irinotecan-induced severe toxicities is related in part to UGT1A1*6 (UGT, UDP glucuronosyltransferase) and UGT1A1*28, variant alleles that reduce the elimination of SN-38, the active metabolite of irinotecan. Irinotecan 200-205 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 94-97 25880011-1 2015 BACKGROUND: In Asians, the risk of irinotecan-induced severe toxicities is related in part to UGT1A1*6 (UGT, UDP glucuronosyltransferase) and UGT1A1*28, variant alleles that reduce the elimination of SN-38, the active metabolite of irinotecan. Irinotecan 200-205 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 94-100 25890497-0 2015 The oncogenic receptor ErbB2 modulates gemcitabine and irinotecan/SN-38 chemoresistance of human pancreatic cancer cells via hCNT1 transporter and multidrug-resistance associated protein MRP-2. Irinotecan 55-65 erb-b2 receptor tyrosine kinase 2 Homo sapiens 23-28 25880011-1 2015 BACKGROUND: In Asians, the risk of irinotecan-induced severe toxicities is related in part to UGT1A1*6 (UGT, UDP glucuronosyltransferase) and UGT1A1*28, variant alleles that reduce the elimination of SN-38, the active metabolite of irinotecan. Irinotecan 232-242 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 94-97 25880011-1 2015 BACKGROUND: In Asians, the risk of irinotecan-induced severe toxicities is related in part to UGT1A1*6 (UGT, UDP glucuronosyltransferase) and UGT1A1*28, variant alleles that reduce the elimination of SN-38, the active metabolite of irinotecan. Irinotecan 232-242 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 94-100 25880011-2 2015 We prospectively studied the relation between the UGT1A1 genotype and the safety of irinotecan-based regimens in Japanese patients with advanced colorectal cancer, and then constructed a nomogram for predicting the risk of severe neutropenia in the first treatment cycle. Irinotecan 84-94 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 50-56 25880011-6 2015 RESULTS: The UGT1A1 genotype was confirmed to be associated with increased risks of irinotecan-induced grade 3 or 4 neutropenia and diarrhoea. Irinotecan 84-94 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 13-19 25890497-0 2015 The oncogenic receptor ErbB2 modulates gemcitabine and irinotecan/SN-38 chemoresistance of human pancreatic cancer cells via hCNT1 transporter and multidrug-resistance associated protein MRP-2. Irinotecan 55-65 solute carrier family 28 member 1 Homo sapiens 125-130 25890497-0 2015 The oncogenic receptor ErbB2 modulates gemcitabine and irinotecan/SN-38 chemoresistance of human pancreatic cancer cells via hCNT1 transporter and multidrug-resistance associated protein MRP-2. Irinotecan 66-71 erb-b2 receptor tyrosine kinase 2 Homo sapiens 23-28 26027741-8 2015 Heritable genetic variants in the ABC and SLC transport pathways; in the CYP450, GST, and UGT-mediated phase I and II metabolism; in the folate metabolic pathway; as well as in the EGF and VEGF signaling pathways, have been associated with a distinct tumor sensitivity phenotype in CRC patients treated with fluoropyrimidines combined with either irinotecan, oxaliplatin or targeted biological agents. Irinotecan 347-357 ATP binding cassette subfamily B member 6 (Langereis blood group) Homo sapiens 34-37 26027741-8 2015 Heritable genetic variants in the ABC and SLC transport pathways; in the CYP450, GST, and UGT-mediated phase I and II metabolism; in the folate metabolic pathway; as well as in the EGF and VEGF signaling pathways, have been associated with a distinct tumor sensitivity phenotype in CRC patients treated with fluoropyrimidines combined with either irinotecan, oxaliplatin or targeted biological agents. Irinotecan 347-357 C-C motif chemokine ligand 21 Homo sapiens 42-45 26027741-8 2015 Heritable genetic variants in the ABC and SLC transport pathways; in the CYP450, GST, and UGT-mediated phase I and II metabolism; in the folate metabolic pathway; as well as in the EGF and VEGF signaling pathways, have been associated with a distinct tumor sensitivity phenotype in CRC patients treated with fluoropyrimidines combined with either irinotecan, oxaliplatin or targeted biological agents. Irinotecan 347-357 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 90-93 26027741-8 2015 Heritable genetic variants in the ABC and SLC transport pathways; in the CYP450, GST, and UGT-mediated phase I and II metabolism; in the folate metabolic pathway; as well as in the EGF and VEGF signaling pathways, have been associated with a distinct tumor sensitivity phenotype in CRC patients treated with fluoropyrimidines combined with either irinotecan, oxaliplatin or targeted biological agents. Irinotecan 347-357 vascular endothelial growth factor A Homo sapiens 189-193 25981652-2 2015 However, there are reports of an association between certain UGT1A1 genetic polymorphisms and the development of adverse reactions(such as neutropenia)related to irinotecan metabolism. Irinotecan 162-172 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 61-67 25981652-7 2015 These results suggest that a dose reduction of irinotecan should be considered for patients who fall into the homozygous group upon analysis of their UGT1A1 genetic polymorphisms, as such patients might be susceptible to grade 4 neutropenia. Irinotecan 47-57 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 150-156 25762723-3 2015 Several drugs are either substrates or inhibitors of multidrug resistance protein 4 (MRP4), such as the anti-colon cancer drug irinotecan and an anti-retroviral used to treat HIV infection, 3"-azido-3"-deoxythymidine (AZT). Irinotecan 127-137 ATP binding cassette subfamily C member 4 Homo sapiens 53-83 25762723-3 2015 Several drugs are either substrates or inhibitors of multidrug resistance protein 4 (MRP4), such as the anti-colon cancer drug irinotecan and an anti-retroviral used to treat HIV infection, 3"-azido-3"-deoxythymidine (AZT). Irinotecan 127-137 ATP binding cassette subfamily C member 4 Homo sapiens 85-89 26634139-0 2015 CPT-11/bevacizumab for the treatment of refractory brain metastases in patients with HER2-neu-positive breast cancer. Irinotecan 0-6 erb-b2 receptor tyrosine kinase 2 Homo sapiens 85-89 25708528-0 2015 Fatty acid synthase-positive hepatocytes and subsequent steatosis in rat livers by irinotecan. Irinotecan 83-93 fatty acid synthase Rattus norvegicus 0-19 25708528-8 2015 Thus, irinotecan-induced liver steatosis was preceded by Fasn-strongly-positive hepatocytes and liver progenitor cell activation. Irinotecan 6-16 fatty acid synthase Rattus norvegicus 57-61 26634139-3 2015 We evaluated CPT-11 and bevacizumab, which can both cross the blood-brain barrier, as combination therapy to treat HER2-neu-positive breast cancer with brain metastases. Irinotecan 13-19 erb-b2 receptor tyrosine kinase 2 Homo sapiens 115-119 25744244-2 2015 This work aimed to investigate the alterations in chemosensitivity of A549 lung cancer cells for 5-Fluorouracil (5-FU) and irinotecan by silencing ABCE1 using specific small interfering RNAs (siRNA). Irinotecan 123-133 ATP binding cassette subfamily E member 1 Homo sapiens 147-152 26097871-4 2015 In vitro, the bioconjugate selectively interacted with ovarian cancer cells through the CD44 receptor, disclosed a dose-dependent tumor growth inhibition efficacy comparable to that of free SN-38 drug, and inhibited Topoisomerase I function leading to apoptosis by a mechanism involving caspase-3 and -7 activation and PARP cleavage. Irinotecan 190-195 CD44 antigen Mus musculus 88-92 25774912-9 2015 In vivo, the BRCA mutant cell line MX-1 treated with CPT-11 alone demonstrated significant decreased tumor growth; this decrease was enhanced further with the addition of ABT-888. Irinotecan 53-59 BRCA1 DNA repair associated Homo sapiens 13-17 25611302-0 2015 OATP1B1 and tumour OATP1B3 modulate exposure, toxicity, and survival after irinotecan-based chemotherapy. Irinotecan 75-85 solute carrier organic anion transporter family member 1B1 Homo sapiens 0-7 25611302-2 2015 The active metabolite of irinotecan, SN-38, is a known substrate of drug-metabolising enzymes, including UGT1A1, as well as OATP and ABC drug transporters. Irinotecan 37-42 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 105-111 25611302-0 2015 OATP1B1 and tumour OATP1B3 modulate exposure, toxicity, and survival after irinotecan-based chemotherapy. Irinotecan 75-85 solute carrier organic anion transporter family member 1B3 Homo sapiens 19-26 25611302-5 2015 RESULTS: SLCO1B1 521C was significantly associated with increased SN-38 exposure (P<0.001), which was additive with UGT1A1*28. Irinotecan 66-71 solute carrier organic anion transporter family member 1B1 Homo sapiens 9-16 25611302-5 2015 RESULTS: SLCO1B1 521C was significantly associated with increased SN-38 exposure (P<0.001), which was additive with UGT1A1*28. Irinotecan 66-71 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 119-125 25611302-2 2015 The active metabolite of irinotecan, SN-38, is a known substrate of drug-metabolising enzymes, including UGT1A1, as well as OATP and ABC drug transporters. Irinotecan 25-35 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 105-111 25611302-10 2015 CONCLUSIONS: Clarifying the association of host genetic variation in OATP and ABC transporters to SN-38 exposure, toxicity and PFS provides rationale for personalising irinotecan-based chemotherapy. Irinotecan 98-103 solute carrier organic anion transporter family member 1A2 Homo sapiens 69-73 25645751-0 2015 Surface-enhanced Raman spectroscopy of the anti-cancer drug irinotecan in presence of human serum albumin. Irinotecan 60-70 albumin Homo sapiens 92-105 25611302-10 2015 CONCLUSIONS: Clarifying the association of host genetic variation in OATP and ABC transporters to SN-38 exposure, toxicity and PFS provides rationale for personalising irinotecan-based chemotherapy. Irinotecan 168-178 solute carrier organic anion transporter family member 1A2 Homo sapiens 69-73 25433165-10 2015 The expression of OATP2A1 increased the sensitivity of AGS cells against irinotecan which led to reduced cell viability. Irinotecan 73-83 solute carrier organic anion transporter family member 2A1 Homo sapiens 18-25 25733708-0 2015 Activity of MM-398, nanoliposomal irinotecan (nal-IRI), in Ewing"s family tumor xenografts is associated with high exposure of tumor to drug and high SLFN11 expression. Irinotecan 34-44 schlafen family member 11 Homo sapiens 150-156 25733708-9 2015 Cytotoxicity of SN-38 inversely correlated with SLFN11 mRNA expression in 20 EFT cell lines. Irinotecan 16-21 schlafen family member 11 Homo sapiens 48-54 25605843-1 2015 PURPOSE: The phase III CRYSTAL study demonstrated that addition of cetuximab to fluorouracil, leucovorin, and irinotecan (FOLFIRI) significantly improved overall survival, progression-free survival, and objective response in the first-line treatment of patients with KRAS codon 12/13 (exon 2) wild-type metastatic colorectal cancer (mCRC). Irinotecan 110-120 KRAS proto-oncogene, GTPase Homo sapiens 267-271 25554586-2 2015 Substrates transported by ABCC2 include antiepileptics, statins, tenofovir, cisplatin, irinotecan, and carbamazepine. Irinotecan 87-97 ATP binding cassette subfamily C member 2 Homo sapiens 26-31 25783977-5 2015 The evidence to support broad utilization of uridine 5"-diphospho-glucuronosyltransferase 1A1 (UGT1A1) and dihydropyrimidine dehydrogenase (DPD) testing to guide irinotecan and fluorouracil dosing remains limited. Irinotecan 162-172 dihydropyrimidine dehydrogenase Homo sapiens 140-143 25601188-0 2015 Favorable response to trastuzumab plus irinotecan combination therapy in two patients with HER2-positive relapsed small-cell lung cancer. Irinotecan 39-49 erb-b2 receptor tyrosine kinase 2 Homo sapiens 91-95 25601188-5 2015 Based on these preclinical data, we treated two patients with HER2-positive SCLC by combination of trastuzumab (6 mg/kg, day 1) and irinotecan (80 mg/m(2), days 1 and 8) every 21 days as the third-line chemotherapy following two prior regimens, first-line carboplatin plus etoposide and second-line amrubicin. Irinotecan 132-142 erb-b2 receptor tyrosine kinase 2 Homo sapiens 62-66 25601188-9 2015 In conclusion, trastuzumab plus irinotecan chemotherapy is promising and feasible against HER2-positive relapsed SCLC. Irinotecan 32-42 erb-b2 receptor tyrosine kinase 2 Homo sapiens 90-94 25644220-1 2015 In this study, we successfully synthesized d-alpha-tocopheryl polyethylene glycol 2000 succinate (TPGS2k) and prepared TPGS2k-modified poly(lactic-co-glycolic acid) nanoparticles (TPGS2k/PLGA NPs) loaded with 7-ethyl-10-hydroxycamptothecin (SN-38), designated TPGS2k/PLGA/SN-38 NPs. Irinotecan 209-239 tubulin polyglutamylase complex subunit 2 Homo sapiens 119-124 25644220-1 2015 In this study, we successfully synthesized d-alpha-tocopheryl polyethylene glycol 2000 succinate (TPGS2k) and prepared TPGS2k-modified poly(lactic-co-glycolic acid) nanoparticles (TPGS2k/PLGA NPs) loaded with 7-ethyl-10-hydroxycamptothecin (SN-38), designated TPGS2k/PLGA/SN-38 NPs. Irinotecan 241-246 tubulin polyglutamylase complex subunit 2 Homo sapiens 119-124 25644220-1 2015 In this study, we successfully synthesized d-alpha-tocopheryl polyethylene glycol 2000 succinate (TPGS2k) and prepared TPGS2k-modified poly(lactic-co-glycolic acid) nanoparticles (TPGS2k/PLGA NPs) loaded with 7-ethyl-10-hydroxycamptothecin (SN-38), designated TPGS2k/PLGA/SN-38 NPs. Irinotecan 272-277 tubulin polyglutamylase complex subunit 2 Homo sapiens 119-124 25644220-3 2015 SN-38 was encapsulated in TPGS2k emulsified PLGA NPs with the entrapment efficiency and loading rates of SN-38 83.6 and 7.85%, respectively. Irinotecan 0-5 tubulin polyglutamylase complex subunit 2 Homo sapiens 26-31 25644220-3 2015 SN-38 was encapsulated in TPGS2k emulsified PLGA NPs with the entrapment efficiency and loading rates of SN-38 83.6 and 7.85%, respectively. Irinotecan 105-110 tubulin polyglutamylase complex subunit 2 Homo sapiens 26-31 25644220-4 2015 SN-38 could release constantly from TPGS2k/PLGA/SN-38 NPs in vitro. Irinotecan 0-5 tubulin polyglutamylase complex subunit 2 Homo sapiens 36-41 25644220-4 2015 SN-38 could release constantly from TPGS2k/PLGA/SN-38 NPs in vitro. Irinotecan 48-53 tubulin polyglutamylase complex subunit 2 Homo sapiens 36-41 25644220-5 2015 TPGS2k/PLGA/SN-38 NPs induced significantly higher cytotoxicity on A549 cells and the multidrug resistance (MDR) cell line (A549/DDP cells and A549/Taxol cells) compared with free SN-38. Irinotecan 12-17 tubulin polyglutamylase complex subunit 2 Homo sapiens 0-5 25644220-5 2015 TPGS2k/PLGA/SN-38 NPs induced significantly higher cytotoxicity on A549 cells and the multidrug resistance (MDR) cell line (A549/DDP cells and A549/Taxol cells) compared with free SN-38. Irinotecan 180-185 tubulin polyglutamylase complex subunit 2 Homo sapiens 0-5 25644220-6 2015 Further studies on the mechanism of the NPs in increasing the death of MDR cells showed that following the SN-38 releasing into cytoplasm the remaining TPGS2k/PLGA NPs could reverse the P-gp mediated MDR via interfering with the structure and function of mitochondria and rather than directly inhibiting the enzymatic activity of P-gp ATPase. Irinotecan 107-112 tubulin polyglutamylase complex subunit 2 Homo sapiens 152-157 25644220-6 2015 Further studies on the mechanism of the NPs in increasing the death of MDR cells showed that following the SN-38 releasing into cytoplasm the remaining TPGS2k/PLGA NPs could reverse the P-gp mediated MDR via interfering with the structure and function of mitochondria and rather than directly inhibiting the enzymatic activity of P-gp ATPase. Irinotecan 107-112 phosphoglycolate phosphatase Homo sapiens 186-190 25644220-6 2015 Further studies on the mechanism of the NPs in increasing the death of MDR cells showed that following the SN-38 releasing into cytoplasm the remaining TPGS2k/PLGA NPs could reverse the P-gp mediated MDR via interfering with the structure and function of mitochondria and rather than directly inhibiting the enzymatic activity of P-gp ATPase. Irinotecan 107-112 phosphoglycolate phosphatase Homo sapiens 330-334 25885574-0 2015 A phase II study of weekly irinotecan in patients with locally advanced or metastatic HER2- negative breast cancer and increased copy numbers of the topoisomerase 1 (TOP1) gene: a study protocol. Irinotecan 27-37 erb-b2 receptor tyrosine kinase 2 Homo sapiens 86-90 25462817-6 2015 Selected compounds were tested for their ability to sensitize MRP4-overexpressing cell lines to the MRP4 substrate drugs 6-mercaptopurine and SN-38, with sensitization up to 6.5-fold with the ryanodine receptor antagonist dantrolene. Irinotecan 142-147 ATP binding cassette subfamily C member 4 Homo sapiens 62-66 25689738-4 2015 In particular, a polymorphism in the UGT1A1 gene (UGT1A1*28) has been linked to an impaired detoxification of SN-38 (irinotecan active metabolite) to SN-38 glucuronide (SN-38G) leading to increased toxicities. Irinotecan 110-115 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 37-43 25611974-0 2015 Thymoquinone induces caspase-independent, autophagic cell death in CPT-11-resistant lovo colon cancer via mitochondrial dysfunction and activation of JNK and p38. Irinotecan 67-73 mitogen-activated protein kinase 14 Homo sapiens 158-161 25713534-3 2015 Hangeshashinto improves irinotecan-induced diarrhea and chemotherapy-induced mucositis by inhibiting the activity of beta-glucuronidase as well as the synthesis of prostaglandin E2. Irinotecan 24-34 glucuronidase beta Homo sapiens 117-135 25685067-0 2015 Treatment with the PARP inhibitor, niraparib, sensitizes colorectal cancer cell lines to irinotecan regardless of MSI/MSS status. Irinotecan 89-99 poly(ADP-ribose) polymerase 1 Homo sapiens 19-23 25689738-4 2015 In particular, a polymorphism in the UGT1A1 gene (UGT1A1*28) has been linked to an impaired detoxification of SN-38 (irinotecan active metabolite) to SN-38 glucuronide (SN-38G) leading to increased toxicities. Irinotecan 110-115 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 50-56 25689738-4 2015 In particular, a polymorphism in the UGT1A1 gene (UGT1A1*28) has been linked to an impaired detoxification of SN-38 (irinotecan active metabolite) to SN-38 glucuronide (SN-38G) leading to increased toxicities. Irinotecan 117-127 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 37-43 25689738-4 2015 In particular, a polymorphism in the UGT1A1 gene (UGT1A1*28) has been linked to an impaired detoxification of SN-38 (irinotecan active metabolite) to SN-38 glucuronide (SN-38G) leading to increased toxicities. Irinotecan 117-127 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 50-56 25689738-4 2015 In particular, a polymorphism in the UGT1A1 gene (UGT1A1*28) has been linked to an impaired detoxification of SN-38 (irinotecan active metabolite) to SN-38 glucuronide (SN-38G) leading to increased toxicities. Irinotecan 150-155 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 37-43 25689738-4 2015 In particular, a polymorphism in the UGT1A1 gene (UGT1A1*28) has been linked to an impaired detoxification of SN-38 (irinotecan active metabolite) to SN-38 glucuronide (SN-38G) leading to increased toxicities. Irinotecan 150-155 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 50-56 25971303-1 2015 INTRODUCTION: Pegylated irinotecan NKTR-102 is a topoisomerase I inhibitor-polymer conjugate. Irinotecan 24-34 natural killer cell triggering receptor Homo sapiens 35-39 25971303-3 2015 To the best of our knowledge, this is the first case report of an impressive iterative response to pegylated irinotecan NKTR-102 in metastatic breast cancer. Irinotecan 109-119 natural killer cell triggering receptor Homo sapiens 120-124 25971303-5 2015 She showed no major response to conventional treatment, whereas, the tumor shrinkage under pegylated irinotecan NKTR-102 was impressive, durable and iterative. Irinotecan 101-111 natural killer cell triggering receptor Homo sapiens 112-116 25462817-6 2015 Selected compounds were tested for their ability to sensitize MRP4-overexpressing cell lines to the MRP4 substrate drugs 6-mercaptopurine and SN-38, with sensitization up to 6.5-fold with the ryanodine receptor antagonist dantrolene. Irinotecan 142-147 ATP binding cassette subfamily C member 4 Homo sapiens 100-104 25522766-0 2015 Clinical and cellular roles for TDP1 and TOP1 in modulating colorectal cancer response to irinotecan. Irinotecan 90-100 tyrosyl-DNA phosphodiesterase 1 Homo sapiens 32-36 25445689-1 2015 The binding of irinotecan to serum proteins (hemoglobin, globulin and human serum albumin) was studied on the surface of epoxide modified superparamagnetic iron oxide nanoparticles (GPTS-SPIONs), which were synthesized by the coprecipitation of ferrous and ferric salts with NH4OH and then modified with [3-(2,3-epoxypropoxy)propyl] trimethoxy silane (GPTS) to obtain functional epoxide groups on the SPIONs" surface. Irinotecan 15-25 albumin Homo sapiens 76-89 25522766-8 2015 Conversely, TDP1 depletion increases DNA-strand breakage and hypersensitivity to irinotecan in a TOP1-dependent manner, presenting a potential therapeutic opportunity in colorectal cancer. Irinotecan 81-91 tyrosyl-DNA phosphodiesterase 1 Homo sapiens 12-16 25055799-0 2015 UGT1A1 *6 polymorphism predicts outcome in elderly patients with relapsed or refractory diffuse large B-cell lymphoma treated with carboplatin, dexamethasone, etoposide and irinotecan. Irinotecan 173-183 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 25055799-1 2015 The uridine diphosphate glucuronosyltransferase (UGT) gene 1A1*6 polymorphism, which affects irinotecan metabolism, has been associated with improved survival in lymphoma patients treated with of carboplatin, dexamethasone, etoposide and irinotecan (CDE-11). Irinotecan 93-103 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 4-47 25055799-1 2015 The uridine diphosphate glucuronosyltransferase (UGT) gene 1A1*6 polymorphism, which affects irinotecan metabolism, has been associated with improved survival in lymphoma patients treated with of carboplatin, dexamethasone, etoposide and irinotecan (CDE-11). Irinotecan 93-103 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 49-52 25055799-1 2015 The uridine diphosphate glucuronosyltransferase (UGT) gene 1A1*6 polymorphism, which affects irinotecan metabolism, has been associated with improved survival in lymphoma patients treated with of carboplatin, dexamethasone, etoposide and irinotecan (CDE-11). Irinotecan 238-248 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 4-47 25055799-1 2015 The uridine diphosphate glucuronosyltransferase (UGT) gene 1A1*6 polymorphism, which affects irinotecan metabolism, has been associated with improved survival in lymphoma patients treated with of carboplatin, dexamethasone, etoposide and irinotecan (CDE-11). Irinotecan 238-248 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 49-52 25466509-2 2015 PATIENTS AND METHODS: The results for efficacy and safety over the time course of the VEGF Trap (aflibercept) with irinotecan in colorectal cancer after failure of oxaliplatin regimen trial were analysed based on data from 1226 patients randomised to receive FOLFIRI plus either aflibercept (n=612) or placebo (n=614). Irinotecan 115-125 vascular endothelial growth factor A Homo sapiens 86-90 25648497-13 2015 In HT-29, HCT-116 and HCT-15 colorectal cancer lines, GHRH antagonist treatment caused cellular arrest in S-phase of cell cycle, potentiated inhibition of in vitro proliferation and in vivo growth produced by S-phase specific cytotoxic agents, 5-FU, irinotecan and cisplatin. Irinotecan 250-260 growth hormone releasing hormone Homo sapiens 54-58 25786458-0 2015 Therapy-related leukemia with Inv(16)(p13.1q22) and type D CBFB/MYH11 developing after exposure to irinotecan-containing chemoradiotherapy. Irinotecan 99-109 inversin Homo sapiens 30-33 25767314-0 2015 Liquid Chromatographic Method for Irinotecan Estimation: Screening of P-gp Modulators. Irinotecan 34-44 phosphoglycolate phosphatase Homo sapiens 70-74 25767314-1 2015 The present work is aimed to develop a simple, sensitive, robust and reliable HPLC method for the estimation of irinotecan in the physiological media in order to assess the permeability profile of irinotecan, using the everted gut sac, in the presence of various P-gp modulators. Irinotecan 112-122 phosphoglycolate phosphatase Homo sapiens 263-267 25767314-7 2015 The developed method was found to be precise (RSD < 1.5%, for repeatability and <2.55% for intermediate precision, acceptable ranges of precision), accurate (The recovered content of irinotecan in the presence of various P-gp modulators varied from 96.11-101.51%, within acceptable range, 80-120%), specific and robust (% RSD < 2). Irinotecan 189-199 phosphoglycolate phosphatase Homo sapiens 227-231 25786458-0 2015 Therapy-related leukemia with Inv(16)(p13.1q22) and type D CBFB/MYH11 developing after exposure to irinotecan-containing chemoradiotherapy. Irinotecan 99-109 core-binding factor subunit beta Homo sapiens 59-63 25786458-0 2015 Therapy-related leukemia with Inv(16)(p13.1q22) and type D CBFB/MYH11 developing after exposure to irinotecan-containing chemoradiotherapy. Irinotecan 99-109 myosin heavy chain 11 Homo sapiens 64-69 25352209-0 2015 Knockdown of Merm1/Wbscr22 attenuates sensitivity of H460 non-small cell lung cancer cells to SN-38 and 5-FU without alteration to p53 expression levels. Irinotecan 94-99 BUD23 rRNA methyltransferase and ribosome maturation factor Homo sapiens 13-18 25236593-0 2015 Erratum to: Effect of simvastatin plus cetuximab/irinotecan for KRAS mutant colorectal cancer and predictive value of the RAS signature for treatment response to cetuximab. Irinotecan 49-59 KRAS proto-oncogene, GTPase Homo sapiens 64-68 25352209-0 2015 Knockdown of Merm1/Wbscr22 attenuates sensitivity of H460 non-small cell lung cancer cells to SN-38 and 5-FU without alteration to p53 expression levels. Irinotecan 94-99 BUD23 rRNA methyltransferase and ribosome maturation factor Homo sapiens 19-26 25352209-7 2015 Downregulation of Merm1/Wbscr22 did not affect H1299 sensitivity to any of the six antitumor agents, whereas attenuated H460 sensitivity to SN-38 and 5-FU, without significant alteration in p53 at both mRNA and protein levels, was identified. Irinotecan 140-145 BUD23 rRNA methyltransferase and ribosome maturation factor Homo sapiens 18-23 25352209-11 2015 In conclusion, shRNA-mediated knockdown of Merm1/Wbscr22 attenuates H460 sensitivity to SN-38 and 5-FU, suggesting Merm1/Wbscr22 is involved in chemosensitivity to SN-38 and 5-FU in H460 cells. Irinotecan 88-93 BUD23 rRNA methyltransferase and ribosome maturation factor Homo sapiens 43-48 25352209-11 2015 In conclusion, shRNA-mediated knockdown of Merm1/Wbscr22 attenuates H460 sensitivity to SN-38 and 5-FU, suggesting Merm1/Wbscr22 is involved in chemosensitivity to SN-38 and 5-FU in H460 cells. Irinotecan 88-93 BUD23 rRNA methyltransferase and ribosome maturation factor Homo sapiens 49-56 25352209-11 2015 In conclusion, shRNA-mediated knockdown of Merm1/Wbscr22 attenuates H460 sensitivity to SN-38 and 5-FU, suggesting Merm1/Wbscr22 is involved in chemosensitivity to SN-38 and 5-FU in H460 cells. Irinotecan 88-93 BUD23 rRNA methyltransferase and ribosome maturation factor Homo sapiens 115-120 25352209-11 2015 In conclusion, shRNA-mediated knockdown of Merm1/Wbscr22 attenuates H460 sensitivity to SN-38 and 5-FU, suggesting Merm1/Wbscr22 is involved in chemosensitivity to SN-38 and 5-FU in H460 cells. Irinotecan 88-93 BUD23 rRNA methyltransferase and ribosome maturation factor Homo sapiens 121-128 25352209-11 2015 In conclusion, shRNA-mediated knockdown of Merm1/Wbscr22 attenuates H460 sensitivity to SN-38 and 5-FU, suggesting Merm1/Wbscr22 is involved in chemosensitivity to SN-38 and 5-FU in H460 cells. Irinotecan 164-169 BUD23 rRNA methyltransferase and ribosome maturation factor Homo sapiens 43-48 25352209-11 2015 In conclusion, shRNA-mediated knockdown of Merm1/Wbscr22 attenuates H460 sensitivity to SN-38 and 5-FU, suggesting Merm1/Wbscr22 is involved in chemosensitivity to SN-38 and 5-FU in H460 cells. Irinotecan 164-169 BUD23 rRNA methyltransferase and ribosome maturation factor Homo sapiens 49-56 25352209-11 2015 In conclusion, shRNA-mediated knockdown of Merm1/Wbscr22 attenuates H460 sensitivity to SN-38 and 5-FU, suggesting Merm1/Wbscr22 is involved in chemosensitivity to SN-38 and 5-FU in H460 cells. Irinotecan 164-169 BUD23 rRNA methyltransferase and ribosome maturation factor Homo sapiens 115-120 25352209-11 2015 In conclusion, shRNA-mediated knockdown of Merm1/Wbscr22 attenuates H460 sensitivity to SN-38 and 5-FU, suggesting Merm1/Wbscr22 is involved in chemosensitivity to SN-38 and 5-FU in H460 cells. Irinotecan 164-169 BUD23 rRNA methyltransferase and ribosome maturation factor Homo sapiens 121-128 25738310-11 2015 Sorafenib and irinotecan sequential treatment significantly increased the levels of cleaved caspase-8, -3, and PARP in HepG2 cells. Irinotecan 14-24 caspase 8 Homo sapiens 92-101 25539916-7 2015 Finally, we provide cellular evidence that LIG3 and not PARP1 acts as the sensor for DNA damage caused by the topoisomerase I inhibitor, irinotecan. Irinotecan 137-147 DNA ligase 3 Homo sapiens 43-47 25738310-11 2015 Sorafenib and irinotecan sequential treatment significantly increased the levels of cleaved caspase-8, -3, and PARP in HepG2 cells. Irinotecan 14-24 collagen type XI alpha 2 chain Homo sapiens 111-115 25738310-12 2015 Sorafenib suppressed p53 expression at both mRNA and protein levels, which might contribute to cell cycle arrest and sensitize tumor cells to irinotecan. Irinotecan 142-152 tumor protein p53 Homo sapiens 21-24 25427841-0 2015 A phase I study of UGT1A1 *28/*6 genotype-directed dosing of irinotecan (CPT-11) in Korean patients with metastatic colorectal cancer receiving FOLFIRI. Irinotecan 61-71 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 19-25 25427841-0 2015 A phase I study of UGT1A1 *28/*6 genotype-directed dosing of irinotecan (CPT-11) in Korean patients with metastatic colorectal cancer receiving FOLFIRI. Irinotecan 73-79 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 19-25 25427841-1 2015 PURPOSE: A UGT1A1 genotype-directed dose escalation of irinotecan (CPT-11) was performed in patients with metastatic colorectal cancer receiving first-line FOLFIRI chemotherapy. Irinotecan 55-65 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 11-17 25565667-0 2015 Irinotecan treatment and senescence failure promote the emergence of more transformed and invasive cells that depend on anti-apoptotic Mcl-1. Irinotecan 0-10 myeloid cell leukemia sequence 1 Mus musculus 135-140 25427841-1 2015 PURPOSE: A UGT1A1 genotype-directed dose escalation of irinotecan (CPT-11) was performed in patients with metastatic colorectal cancer receiving first-line FOLFIRI chemotherapy. Irinotecan 67-73 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 11-17 25565667-9 2015 Depletion of Mcl-1 increased irinotecan efficiency, induced the death of polyploid cells, prevented cell emergence and inhibited growth in low-adhesion conditions. Irinotecan 29-39 myeloid cell leukemia sequence 1 Mus musculus 13-18 25565667-10 2015 We therefore propose that Mcl-1 targeting should be considered in the future to reduce senescence escape and to improve the treatment of irinotecan-refractory colorectal cancers. Irinotecan 137-147 myeloid cell leukemia sequence 1 Mus musculus 26-31 25427841-8 2015 CONCLUSIONS: The MTD of irinotecan differed according to the UGT1A1 genotype, and higher doses of irinotecan are feasible with sLV5FU2 compared to the present regulatory approved doses. Irinotecan 24-34 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 61-67 25427841-8 2015 CONCLUSIONS: The MTD of irinotecan differed according to the UGT1A1 genotype, and higher doses of irinotecan are feasible with sLV5FU2 compared to the present regulatory approved doses. Irinotecan 98-108 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 61-67 25481222-5 2015 Whereas NTCP did not show any strong interactions with the cytostatics tested, we observed a very strong inhibition of OAT2 mediated [(3)H] cGMP uptake in the presence of bendamustine, irinotecan and paclitaxel. Irinotecan 185-195 solute carrier family 22 member 7 Homo sapiens 119-123 25481222-6 2015 The Ki values of OAT2 for bendamustine, irinotecan and paclitaxel were determined to be 43.3+-4.33muM, 26.4+-2.34muM and 10.4+-0.45muM, respectively. Irinotecan 40-50 solute carrier family 22 member 7 Homo sapiens 17-21 25481222-8 2015 A higher accumulation of irinotecan was observed in OAT2 expressing cells compared to control pcDNA cells by HPLC analysis of cell lysates. Irinotecan 25-35 solute carrier family 22 member 7 Homo sapiens 52-56 25481222-9 2015 The accumulation was diminished in the presence of cGMP, the substrate we used to functionally characterize OAT2, suggesting specificity of this uptake and the fact that OAT2 mediates uptake of irinotecan. Irinotecan 194-204 solute carrier family 22 member 7 Homo sapiens 108-112 25481222-9 2015 The accumulation was diminished in the presence of cGMP, the substrate we used to functionally characterize OAT2, suggesting specificity of this uptake and the fact that OAT2 mediates uptake of irinotecan. Irinotecan 194-204 solute carrier family 22 member 7 Homo sapiens 170-174 25833236-2 2015 The present study investigated the anticancer effect of irinotecan on p53-negative Caco-2 and p53-positive CW2 human colorectal cancer cell lines. Irinotecan 56-66 tumor protein p53 Homo sapiens 70-73 26068529-2 2015 UDP-glucuronosyltransferase 1A1 (UGT1A1) plays an irreplaceable role in detoxification of bilirubin and many drugs (e.g., SN-38). Irinotecan 122-127 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-31 26087609-0 2015 [The role of assessing UGT1A1 gene polymorphism in the prediction of irinotecan-induced toxicity in the course of chemotherapy for colorectal cancer]. Irinotecan 69-79 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 23-29 26068529-2 2015 UDP-glucuronosyltransferase 1A1 (UGT1A1) plays an irreplaceable role in detoxification of bilirubin and many drugs (e.g., SN-38). Irinotecan 122-127 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-39 25877315-0 2015 [Association of UGT1A1 (*28, *60 and * 93) polymorphism with the adverse reactions of irinotecan chemotherapy in extensive stage small cell lung cancer]. Irinotecan 86-96 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 16-22 25877315-1 2015 OBJECTIVE: To explore the correlation between UGT1A1 (*28, *60 and * 93) polymorphism and the adverse reactions in small cell lung cancer patients after irinotecan chemotherapy. Irinotecan 153-163 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 46-52 25877315-9 2015 Further prospective study in a larger number of patients is needed to clarify the association between UGT1A1*28, UGT1A1* 93 and UGT1A1*60 polymorphism and adverse reactions of irinotecan, and to help clinicians in choosing a better therapeutic modality for personalized chemotherapy to improve curative effect and reduce adverse reactions. Irinotecan 176-186 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 102-108 24588516-4 2014 As irinotecan is a P-gp substrate, we tested here whether cetuximab could modify irinotecan concentration in mice. Irinotecan 3-13 phosphoglycolate phosphatase Mus musculus 19-23 25474134-0 2014 MBL-II-141, a chromone derivative, enhances irinotecan (CPT-11) anticancer efficiency in ABCG2-positive xenografts. Irinotecan 44-54 ATP binding cassette subfamily G member 2 (Junior blood group) Mus musculus 89-94 25474134-0 2014 MBL-II-141, a chromone derivative, enhances irinotecan (CPT-11) anticancer efficiency in ABCG2-positive xenografts. Irinotecan 56-62 ATP binding cassette subfamily G member 2 (Junior blood group) Mus musculus 89-94 25474134-4 2014 Combination of 10 mg/kg MBL-II-141 with the anticancer agent CPT-11 completely blocked the growth of 90% freshly implanted ABCG2-positive tumors. Irinotecan 61-67 ATP binding cassette subfamily G member 2 (Junior blood group) Mus musculus 123-128 25474134-9 2014 MBL-II-141 induced a potent sensitization of ABCG2-positive xenografts to CPT-11 through in vivo ABCG2 inhibition. Irinotecan 74-80 ATP binding cassette subfamily G member 2 (Junior blood group) Mus musculus 45-50 25474134-9 2014 MBL-II-141 induced a potent sensitization of ABCG2-positive xenografts to CPT-11 through in vivo ABCG2 inhibition. Irinotecan 74-80 ATP binding cassette subfamily G member 2 (Junior blood group) Mus musculus 97-102 25544747-7 2014 In the primary lung cancer mouse models, tumor mass was 80% lower in response to HB1.F3.CE in conjunction with CPT-11, while it was only reduced by 40% in the group treated with CPT-11 alone. Irinotecan 111-117 paternally expressed 10 Mus musculus 81-84 25544747-7 2014 In the primary lung cancer mouse models, tumor mass was 80% lower in response to HB1.F3.CE in conjunction with CPT-11, while it was only reduced by 40% in the group treated with CPT-11 alone. Irinotecan 178-184 paternally expressed 10 Mus musculus 81-84 25515240-0 2014 DEK is a potential marker for aggressive phenotype and irinotecan-based therapy response in metastatic colorectal cancer. Irinotecan 55-65 DEK proto-oncogene Homo sapiens 0-3 25515240-8 2014 DEK expression was determined by immunohistochemistry in 67 formalin-fixed paraffin-embedded tumour samples from metastatic colorectal cancer patients treated with irinotecan-based therapy as first-line treatment. Irinotecan 164-174 DEK proto-oncogene Homo sapiens 0-3 25515240-10 2014 Knock-down of DEK on DLD1 and SW620 cell lines decreased cell migration and increased irinotecan-induced apoptosis. Irinotecan 86-96 DEK proto-oncogene Homo sapiens 14-17 25515240-11 2014 In addition, low DEK expression level predicted irinotecan-based chemotherapy response in metastatic colorectal cancer patients with KRAS wild-type. Irinotecan 48-58 DEK proto-oncogene Homo sapiens 17-20 25515240-11 2014 In addition, low DEK expression level predicted irinotecan-based chemotherapy response in metastatic colorectal cancer patients with KRAS wild-type. Irinotecan 48-58 KRAS proto-oncogene, GTPase Homo sapiens 133-137 25515240-12 2014 CONCLUSIONS: These data suggest DEK overexpression as a crucial event for the emergence of an aggressive phenotype in colorectal cancer and its potential role as biomarker for irinotecan response in those patients with KRAS wild-type status. Irinotecan 176-186 DEK proto-oncogene Homo sapiens 32-35 25515240-12 2014 CONCLUSIONS: These data suggest DEK overexpression as a crucial event for the emergence of an aggressive phenotype in colorectal cancer and its potential role as biomarker for irinotecan response in those patients with KRAS wild-type status. Irinotecan 176-186 KRAS proto-oncogene, GTPase Homo sapiens 219-223 24619075-0 2014 Comment on "KRAS-mutated plasma DNA as predictor of outcome from irinotecan monotherapy in metastatic colorectal cancer". Irinotecan 65-75 KRAS proto-oncogene, GTPase Homo sapiens 12-16 24619079-0 2014 Response to comment on "KRAS-mutated plasma DNA as predictor of outcome from irinotecan monotherapy in metastatic colorectal cancer". Irinotecan 77-87 KRAS proto-oncogene, GTPase Homo sapiens 24-28 25202035-1 2014 OBJECTIVE: To report a case of systemic irinotecan toxicity following regional transarterial chemoembolization with drug-eluting beads loaded with irinotecan (DEBIRI-TACE) in a patient later found to have a homozygous mutation for UGT1A1*28. Irinotecan 40-50 ADAM metallopeptidase domain 17 Homo sapiens 166-170 25202035-1 2014 OBJECTIVE: To report a case of systemic irinotecan toxicity following regional transarterial chemoembolization with drug-eluting beads loaded with irinotecan (DEBIRI-TACE) in a patient later found to have a homozygous mutation for UGT1A1*28. Irinotecan 40-50 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 231-237 25202035-1 2014 OBJECTIVE: To report a case of systemic irinotecan toxicity following regional transarterial chemoembolization with drug-eluting beads loaded with irinotecan (DEBIRI-TACE) in a patient later found to have a homozygous mutation for UGT1A1*28. Irinotecan 147-157 ADAM metallopeptidase domain 17 Homo sapiens 166-170 25202035-1 2014 OBJECTIVE: To report a case of systemic irinotecan toxicity following regional transarterial chemoembolization with drug-eluting beads loaded with irinotecan (DEBIRI-TACE) in a patient later found to have a homozygous mutation for UGT1A1*28. Irinotecan 147-157 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 231-237 25202035-7 2014 Patients with genetic polymorphisms of the genes encoding for the enzyme UGT1A1 may have increased incidence of irinotecan-associated toxicities because of decreased clearance of the active metabolite SN38 via the glucuronidation pathway. Irinotecan 112-122 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 73-79 24588516-7 2014 Those results suggest that cetuximab influence irinotecan distribution into tissues probably due to inhibition of P-gp. Irinotecan 47-57 ATP binding cassette subfamily B member 1 Homo sapiens 114-118 24588516-9 2014 Inhibiting SN-38 efflux by P-gp drug transporters in biliary system and tumour can lead to pharmacokinetic modification and a higher anticancer efficacy. Irinotecan 11-16 ATP binding cassette subfamily B member 1 Homo sapiens 27-31 25473295-0 2014 FOLFIRI and regorafenib combination therapy with dose escalation of irinotecan as fourth-line treatment for patients with metastatic colon cancer according to UGT1A1 genotyping. Irinotecan 68-78 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 159-165 25474278-9 2014 The post-progression survival (PPS) of p53-positive patients undergoing 2nd and/or 3rd line chemotherapy with irinotecan and/or cetuximab was significantly longer compared to p53-negative patients. Irinotecan 110-120 tumor protein p53 Homo sapiens 39-42 25474278-11 2014 mCRC patients with p53 overexpression undergoing an irinotecan containing second- or third-line chemotherapy after oxaliplatin failure have a significantly longer post-progression survival compared to patients without p53 overexpression. Irinotecan 52-62 tumor protein p53 Homo sapiens 19-22 25474278-12 2014 To validate the clinical impact of p53 in patients with mCRC treated with irinotecan- and/or cetuximab further studies are needed. Irinotecan 74-84 tumor protein p53 Homo sapiens 35-38 25473295-1 2014 Here we report a case of metastatic colon cancer treated with 5-fluorouracil, leucovorin, and escalated doses of irinotecan (FOLFIRI) combined with regorafenib in the fourth-line setting after uridine diphosphate glucuronosyltransferase (UGT)1A1 genotyping analysis. Irinotecan 113-123 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 238-245 25473295-9 2014 FOLFIRI, with dose escalation of irinotecan according to UGT1A1 genotyping plus regorafenib appears to be a promising salvage therapy for patients with refractory metastatic colorectal cancer. Irinotecan 33-43 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 57-63 25520880-1 2014 We have previous found a positive correlation between post-therapy TCR repertoire normalization and remission of colorectal cancer (CRC) patients following fluorouracil, leucovorin, and irinotecan (FOLFIRI) plus bevacizumab or Rh-endostatin therapy. Irinotecan 186-196 T cell receptor beta variable 20/OR9-2 (non-functional) Homo sapiens 67-70 25089570-0 2014 Schlafen-11 sensitizes colorectal carcinoma cells to irinotecan. Irinotecan 53-63 schlafen family member 11 Homo sapiens 0-11 25437539-4 2014 PARPi-induced cytotoxicity in EWS cells was 10- to 1,000-fold higher after administration of the DNA-damaging agents irinotecan or temozolomide. Irinotecan 117-127 EWS RNA binding protein 1 Homo sapiens 30-33 25369798-12 2014 Additionally, the use of anti-EGFR MoAbs significantly increased the risk of severe infections when used in conjunction with cisplatin (RR 1.48, 95%CI 1.22 to 1.79, P<0.001) or irinotecan (RR 1.53, 95%CI 1.12 to 2.10, P=0.008). Irinotecan 180-190 epidermal growth factor receptor Homo sapiens 30-34 25089570-4 2014 The present study aimed to investigate the mechanism of SLFN11 in the response of CRC cell lines to SN-38 (an active CPT-11 metabolite) treatment. Irinotecan 100-105 schlafen family member 11 Homo sapiens 56-62 25089570-4 2014 The present study aimed to investigate the mechanism of SLFN11 in the response of CRC cell lines to SN-38 (an active CPT-11 metabolite) treatment. Irinotecan 117-123 schlafen family member 11 Homo sapiens 56-62 25089570-7 2014 The effects of SN-38 treatment on CRC cells with different SLFN11 expression levels were detected, including inhibition of cell growth, induction of apoptosis, and cell cycle arrest. Irinotecan 15-20 schlafen family member 11 Homo sapiens 59-65 25089570-8 2014 This study showed that SLFN11 expression varied between the CRC cell lines and high-level SLFN11 expression promoted SN-38-induced antiproliferative activity, apoptosis, and cell cycle arrest. Irinotecan 117-122 schlafen family member 11 Homo sapiens 23-29 25368234-0 2014 Transcriptional targeting of human liver carboxylesterase (hCE1m6) and simultaneous expression of anti-BCRP shRNA enhances sensitivity of breast cancer cells to CPT-11. Irinotecan 161-167 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 103-107 25089570-8 2014 This study showed that SLFN11 expression varied between the CRC cell lines and high-level SLFN11 expression promoted SN-38-induced antiproliferative activity, apoptosis, and cell cycle arrest. Irinotecan 117-122 schlafen family member 11 Homo sapiens 90-96 25368234-2 2014 We hypothesized that conversion of camptothecin-11 (CPT-11) to its highly cytotoxic metabolite SN-38 by a mutant human carboxyl esterase (hCE1m6) specifically in cancer cells and inhibition of ABCG2 by anti-ABCG2 short hairpin RNA, leads to accumulation of a higher concentration of SN-38, resulting in higher therapeutic efficacy and less toxicity to normal cells. Irinotecan 35-50 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 193-198 25368234-2 2014 We hypothesized that conversion of camptothecin-11 (CPT-11) to its highly cytotoxic metabolite SN-38 by a mutant human carboxyl esterase (hCE1m6) specifically in cancer cells and inhibition of ABCG2 by anti-ABCG2 short hairpin RNA, leads to accumulation of a higher concentration of SN-38, resulting in higher therapeutic efficacy and less toxicity to normal cells. Irinotecan 35-50 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 207-212 25089570-9 2014 Our results suggest that SLFN11 plays a key role in cell cycle arrest and/or induction of apoptosis in response to exogenous SN-38-induced DNA damage and might be used as a new predictive biomarker for CRC treatment. Irinotecan 125-130 schlafen family member 11 Homo sapiens 25-31 25368234-2 2014 We hypothesized that conversion of camptothecin-11 (CPT-11) to its highly cytotoxic metabolite SN-38 by a mutant human carboxyl esterase (hCE1m6) specifically in cancer cells and inhibition of ABCG2 by anti-ABCG2 short hairpin RNA, leads to accumulation of a higher concentration of SN-38, resulting in higher therapeutic efficacy and less toxicity to normal cells. Irinotecan 52-58 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 193-198 25368234-2 2014 We hypothesized that conversion of camptothecin-11 (CPT-11) to its highly cytotoxic metabolite SN-38 by a mutant human carboxyl esterase (hCE1m6) specifically in cancer cells and inhibition of ABCG2 by anti-ABCG2 short hairpin RNA, leads to accumulation of a higher concentration of SN-38, resulting in higher therapeutic efficacy and less toxicity to normal cells. Irinotecan 52-58 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 207-212 25285015-0 2014 Clinical significance of UGT1A1 gene polymorphisms on irinotecan-based regimens as the treatment in metastatic colorectal cancer. Irinotecan 54-64 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 25-31 25368234-2 2014 We hypothesized that conversion of camptothecin-11 (CPT-11) to its highly cytotoxic metabolite SN-38 by a mutant human carboxyl esterase (hCE1m6) specifically in cancer cells and inhibition of ABCG2 by anti-ABCG2 short hairpin RNA, leads to accumulation of a higher concentration of SN-38, resulting in higher therapeutic efficacy and less toxicity to normal cells. Irinotecan 95-100 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 193-198 25368234-2 2014 We hypothesized that conversion of camptothecin-11 (CPT-11) to its highly cytotoxic metabolite SN-38 by a mutant human carboxyl esterase (hCE1m6) specifically in cancer cells and inhibition of ABCG2 by anti-ABCG2 short hairpin RNA, leads to accumulation of a higher concentration of SN-38, resulting in higher therapeutic efficacy and less toxicity to normal cells. Irinotecan 95-100 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 207-212 25450281-1 2014 OBJECTIVE: The aim was to investigate the association between uridine diphosphate glucuronide transferase 1A1 (UGT1A1) gene promoter region polymorphism and irinotecan-related adverse effects and efficacy on recurrent and refractory small cell lung cancer (SCLC). Irinotecan 157-167 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 62-109 25450281-1 2014 OBJECTIVE: The aim was to investigate the association between uridine diphosphate glucuronide transferase 1A1 (UGT1A1) gene promoter region polymorphism and irinotecan-related adverse effects and efficacy on recurrent and refractory small cell lung cancer (SCLC). Irinotecan 157-167 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 111-117 25450281-4 2014 DNA was extracted from peripheral blood and direct sequencing method was employed to test UGT1A1FNx0128 polymorphism, thus analyzing the correlation between UGT1A1FNx0128 polymorphism and irinotecan-related side-effects and efficacy. Irinotecan 188-198 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 157-163 25285015-14 2014 CONCLUSION: For irinotecan-based regimens in metastatic colorectal cancer, the UGT1A1*28 and UGT1A1*6 locus mutations can be regarded as predictors for irinotecan-associated severe delayed diarrhea, whereas no association between UGT1A1 gene polymorphisms and severe neutropenia was observed. Irinotecan 152-162 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-99 25454761-5 2014 Compritol ATO 888 and capmule MCM C8 hybrid lipid mix was employed to prepare irinotecan containing nanostructured lipid carrier (NLC) by using functional excipients with P-gp inhibition activity. Irinotecan 78-88 ATP binding cassette subfamily B member 1 Homo sapiens 171-175 25731272-8 2014 Treatment was changed to LV, 5-fluorouracil, and irinotecan (FOLFIRI), but after 4 courses, the patient"s carcinoembryonic antigen (CEA) levels rose. Irinotecan 49-59 CEA cell adhesion molecule 3 Homo sapiens 106-130 25195021-10 2014 In addition, the method has been successfully applied to determine the intracellular accumulation of SN-38 investigating the transport through ABCB1 (P-gp) and ABCG2 (BCRP) efflux pumps in colorectal cancer cell lines. Irinotecan 101-106 ATP binding cassette subfamily B member 1 Homo sapiens 143-148 25195021-10 2014 In addition, the method has been successfully applied to determine the intracellular accumulation of SN-38 investigating the transport through ABCB1 (P-gp) and ABCG2 (BCRP) efflux pumps in colorectal cancer cell lines. Irinotecan 101-106 phosphoglycolate phosphatase Homo sapiens 150-154 25195021-10 2014 In addition, the method has been successfully applied to determine the intracellular accumulation of SN-38 investigating the transport through ABCB1 (P-gp) and ABCG2 (BCRP) efflux pumps in colorectal cancer cell lines. Irinotecan 101-106 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 160-165 25195021-10 2014 In addition, the method has been successfully applied to determine the intracellular accumulation of SN-38 investigating the transport through ABCB1 (P-gp) and ABCG2 (BCRP) efflux pumps in colorectal cancer cell lines. Irinotecan 101-106 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 167-171 25310185-11 2014 In subgroup analyses, dMRE11 patients treated with irinotecan exhibited unexplained short-term mortality. Irinotecan 51-61 meiotic recombination 11 Drosophila melanogaster 22-28 25272957-6 2014 The benefit on DFS in AREG-/EREG- patients was even stronger in the group that received 5-FU/FA/irinotecan as adjuvant treatment (p=0.002). Irinotecan 96-106 amphiregulin Homo sapiens 22-26 25272957-6 2014 The benefit on DFS in AREG-/EREG- patients was even stronger in the group that received 5-FU/FA/irinotecan as adjuvant treatment (p=0.002). Irinotecan 96-106 epiregulin Homo sapiens 28-32 25272957-7 2014 Patients with strong expression of PTEN profited more in terms of OS under adjuvant treatment containing irinotecan (p< 0.05). Irinotecan 105-115 phosphatase and tensin homolog Homo sapiens 35-39 25272957-14 2014 Nevertheless, AREG/EREG negative, PTEN positive and Hif-1 alpha negative patients might profit significantly in terms of DFS from a treatment containing fluoropyrimidines and irinotecan. Irinotecan 175-185 amphiregulin Homo sapiens 14-18 25272957-14 2014 Nevertheless, AREG/EREG negative, PTEN positive and Hif-1 alpha negative patients might profit significantly in terms of DFS from a treatment containing fluoropyrimidines and irinotecan. Irinotecan 175-185 epiregulin Homo sapiens 19-23 25272957-14 2014 Nevertheless, AREG/EREG negative, PTEN positive and Hif-1 alpha negative patients might profit significantly in terms of DFS from a treatment containing fluoropyrimidines and irinotecan. Irinotecan 175-185 phosphatase and tensin homolog Homo sapiens 34-38 25272957-14 2014 Nevertheless, AREG/EREG negative, PTEN positive and Hif-1 alpha negative patients might profit significantly in terms of DFS from a treatment containing fluoropyrimidines and irinotecan. Irinotecan 175-185 hypoxia inducible factor 1 subunit alpha Homo sapiens 52-63 25064805-5 2014 The combination of Nano Se and irinotecan showed increased cytotoxic effect with HCT-8 tumor cells likely by p53 mediated apoptosis. Irinotecan 31-41 tumor protein p53 Homo sapiens 109-112 25060396-0 2014 Contribution of transcription factor, SP1, to the promotion of HB-EGF expression in defense mechanism against the treatment of irinotecan in ovarian clear cell carcinoma. Irinotecan 127-137 heparin binding EGF like growth factor Homo sapiens 63-69 25175642-0 2014 A novel system for predicting the toxicity of irinotecan based on statistical pattern recognition with UGT1A genotypes. Irinotecan 46-56 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 103-108 24709690-0 2014 The effect of the UGT1A1*28 allele on survival after irinotecan-based chemotherapy: a collaborative meta-analysis. Irinotecan 53-63 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 18-24 24709690-1 2014 To date, studies of irinotecan pharmacogenetics have mostly focused on the effect of the UGT1A1*28 allele on irinotecan-related toxicity. Irinotecan 20-30 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 89-95 24709690-1 2014 To date, studies of irinotecan pharmacogenetics have mostly focused on the effect of the UGT1A1*28 allele on irinotecan-related toxicity. Irinotecan 109-119 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 89-95 24709690-2 2014 However, the clinical utility of routine UGT1A1*28 genotyping to pre-emptively adjust irinotecan dosage is dependent upon whether UGT1A1*28 also affects patient survival following irinotecan therapy. Irinotecan 86-96 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 41-47 24709690-2 2014 However, the clinical utility of routine UGT1A1*28 genotyping to pre-emptively adjust irinotecan dosage is dependent upon whether UGT1A1*28 also affects patient survival following irinotecan therapy. Irinotecan 180-190 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 130-136 24709690-4 2014 A systematic review and meta-analysis of both published and unpublished data were performed to summarize the available evidence of the relationship between the UGT1A1*28 allele and patient survival related to irinotecan therapy. Irinotecan 209-219 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 160-166 25172788-9 2014 Moreover, both cryptotanshinone and dihydrotanshinone could potentiate the cytotoxicity of doxorubicin and irinotecan in P-gp overexpressing SW620 Ad300 colon cancer cells. Irinotecan 107-117 phosphoglycolate phosphatase Homo sapiens 121-125 25285015-1 2014 PURPOSE: The primary aim of this research was to investigate the association between uridine diphosphate glucuronosyltransferase (UGT)1A1 gene polymorphisms and the toxicities of irinotecan-based regimens in Chinese patients with metastatic colorectal cancer. Irinotecan 179-189 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 130-137 25285015-14 2014 CONCLUSION: For irinotecan-based regimens in metastatic colorectal cancer, the UGT1A1*28 and UGT1A1*6 locus mutations can be regarded as predictors for irinotecan-associated severe delayed diarrhea, whereas no association between UGT1A1 gene polymorphisms and severe neutropenia was observed. Irinotecan 16-26 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 79-85 25285015-14 2014 CONCLUSION: For irinotecan-based regimens in metastatic colorectal cancer, the UGT1A1*28 and UGT1A1*6 locus mutations can be regarded as predictors for irinotecan-associated severe delayed diarrhea, whereas no association between UGT1A1 gene polymorphisms and severe neutropenia was observed. Irinotecan 16-26 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-99 25285015-14 2014 CONCLUSION: For irinotecan-based regimens in metastatic colorectal cancer, the UGT1A1*28 and UGT1A1*6 locus mutations can be regarded as predictors for irinotecan-associated severe delayed diarrhea, whereas no association between UGT1A1 gene polymorphisms and severe neutropenia was observed. Irinotecan 16-26 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-99 25285015-14 2014 CONCLUSION: For irinotecan-based regimens in metastatic colorectal cancer, the UGT1A1*28 and UGT1A1*6 locus mutations can be regarded as predictors for irinotecan-associated severe delayed diarrhea, whereas no association between UGT1A1 gene polymorphisms and severe neutropenia was observed. Irinotecan 152-162 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 79-85 25285015-14 2014 CONCLUSION: For irinotecan-based regimens in metastatic colorectal cancer, the UGT1A1*28 and UGT1A1*6 locus mutations can be regarded as predictors for irinotecan-associated severe delayed diarrhea, whereas no association between UGT1A1 gene polymorphisms and severe neutropenia was observed. Irinotecan 152-162 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-99 24934408-4 2014 Whole-genome sequencing revealed a clonal hemizygous mutation in the Mre11 complex gene RAD50 that attenuated ATM signaling which in the context of CHK1 inhibition contributed, via synthetic lethality, to extreme sensitivity to irinotecan. Irinotecan 228-238 MRE11 homolog, double strand break repair nuclease Homo sapiens 69-74 25202056-0 2014 Expression of ABCB6 is related to resistance to 5-FU, SN-38 and vincristine. Irinotecan 54-59 ATP binding cassette subfamily B member 6 (Langereis blood group) Homo sapiens 14-19 25202056-5 2014 ABCB6 expression enhanced resistance to 5-fluorouracil (5-FU), SN-38 and vincristine (Vcr) but not to arsenite. Irinotecan 63-68 ATP binding cassette subfamily B member 6 (Langereis blood group) Homo sapiens 0-5 25202056-6 2014 Conversely, down-regulation of ABCB6 in KAS cells increased the sensitivity of KAS cells to 5-FU, SN-38 and Vcr, but not to arsenite. Irinotecan 98-103 ATP binding cassette subfamily B member 6 (Langereis blood group) Homo sapiens 31-36 25202056-7 2014 Our findings suggest that ABCB6 is involved in 5-FU, SN-38 and Vcr resistance. Irinotecan 53-58 ATP binding cassette subfamily B member 6 (Langereis blood group) Homo sapiens 26-31 25260839-0 2014 Influence of UGT1A1 gene methylation level in colorectal cancer cells on the sensitivity of the chemotherapy drug CPT-11. Irinotecan 114-120 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 13-19 25260839-1 2014 OBJECTIVE: To study the influence of the methylation level of UGT1A1 gene related to CPT-11 metabolic enzymes in colorectal cancer cells on the sensitivity of chemotherapy drugs. Irinotecan 85-91 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 62-68 25260839-6 2014 CONCLUSIONS: The cytotoxicity of CPT-11 to colorectal cancer cells has a negative correlation with UGT1A1 expression, and positive correlation with CES2 and GUSB. Irinotecan 33-39 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 99-105 25260839-6 2014 CONCLUSIONS: The cytotoxicity of CPT-11 to colorectal cancer cells has a negative correlation with UGT1A1 expression, and positive correlation with CES2 and GUSB. Irinotecan 33-39 carboxylesterase 2 Homo sapiens 148-152 25260839-6 2014 CONCLUSIONS: The cytotoxicity of CPT-11 to colorectal cancer cells has a negative correlation with UGT1A1 expression, and positive correlation with CES2 and GUSB. Irinotecan 33-39 glucuronidase beta Homo sapiens 157-161 25260839-7 2014 The specific silencing UGT1A1 gene of siRNA could significantly increase the sensitivity of CPT-11 to the chemotherapy of colorectal cancer cells. Irinotecan 92-98 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 23-29 25260839-8 2014 UGT1A1 methylation was an important factor affecting the chemosensitivity of CPT-11. Irinotecan 77-83 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 25185187-1 2014 SUMMARY: In this issue of Cancer Discovery, AI-Ahmadie and colleagues identify a somatic mutation in the RAD50 gene as a likely contributing factor to an unusual curative response to systemic combination therapy employing the DNA-damaging agent irinotecan and a checkpoint kinase 1 inhibitor in a patient with recurrent, metastatic small-cell cancer. Irinotecan 245-255 RAD50 double strand break repair protein Homo sapiens 105-110 24934408-4 2014 Whole-genome sequencing revealed a clonal hemizygous mutation in the Mre11 complex gene RAD50 that attenuated ATM signaling which in the context of CHK1 inhibition contributed, via synthetic lethality, to extreme sensitivity to irinotecan. Irinotecan 228-238 RAD50 double strand break repair protein Homo sapiens 88-93 24934408-4 2014 Whole-genome sequencing revealed a clonal hemizygous mutation in the Mre11 complex gene RAD50 that attenuated ATM signaling which in the context of CHK1 inhibition contributed, via synthetic lethality, to extreme sensitivity to irinotecan. Irinotecan 228-238 ATM serine/threonine kinase Homo sapiens 110-113 24934408-4 2014 Whole-genome sequencing revealed a clonal hemizygous mutation in the Mre11 complex gene RAD50 that attenuated ATM signaling which in the context of CHK1 inhibition contributed, via synthetic lethality, to extreme sensitivity to irinotecan. Irinotecan 228-238 checkpoint kinase 1 Homo sapiens 148-152 24513691-7 2014 This prospective study did not confirm any previous retrospective finding, reporting a predictive or prognostic effect of EGFR (CA)n repeats allelic variants in chemo-refractory mCRC patients receiving cetuximab and irinotecan. Irinotecan 216-226 epidermal growth factor receptor Homo sapiens 122-126 24424522-2 2014 Here we show that interleukin-33 (IL-33) mediates the severe intestinal mucositis in mice treated with irinotecan (CPT-11), a commonly used cancer chemotherapeutic agent. Irinotecan 103-113 interleukin 33 Mus musculus 34-39 24424522-2 2014 Here we show that interleukin-33 (IL-33) mediates the severe intestinal mucositis in mice treated with irinotecan (CPT-11), a commonly used cancer chemotherapeutic agent. Irinotecan 115-121 interleukin 33 Mus musculus 18-32 24424522-2 2014 Here we show that interleukin-33 (IL-33) mediates the severe intestinal mucositis in mice treated with irinotecan (CPT-11), a commonly used cancer chemotherapeutic agent. Irinotecan 115-121 interleukin 33 Mus musculus 34-39 24424522-3 2014 Systemic CPT-11 administration led to severe mucosal damage, diarrhea, and body weight loss concomitant with the induction of IL-33 in the small intestine (SI). Irinotecan 9-15 interleukin 33 Mus musculus 126-131 24424522-5 2014 Moreover, recombinant IL-33 exacerbated the CPT-11-induced mucositis, whereas IL-33 blockade with anti-IL-33 antibody or soluble ST2 markedly attenuated the disease. Irinotecan 44-50 interleukin 33 Mus musculus 22-27 24424522-7 2014 Supernatants from SI explants treated with CPT-11 enhanced transmigration of neutrophils in vitro in an IL-33-, CXCL1/2-, and CXCR2-dependent manner. Irinotecan 43-49 interleukin 33 Mus musculus 104-109 24424522-7 2014 Supernatants from SI explants treated with CPT-11 enhanced transmigration of neutrophils in vitro in an IL-33-, CXCL1/2-, and CXCR2-dependent manner. Irinotecan 43-49 chemokine (C-X-C motif) ligand 1 Mus musculus 112-117 24424522-7 2014 Supernatants from SI explants treated with CPT-11 enhanced transmigration of neutrophils in vitro in an IL-33-, CXCL1/2-, and CXCR2-dependent manner. Irinotecan 43-49 chemokine (C-X-C motif) receptor 2 Mus musculus 126-131 24424522-8 2014 Importantly, IL-33 blockade reduced mucositis and enabled prolonged CPT-11 treatment of ectopic CT26 colon carcinoma, leading to a beneficial outcome of the chemotherapy. Irinotecan 68-74 interleukin 33 Mus musculus 13-18 25127363-6 2014 We identified the SNP rs9351963 in potassium voltage-gated channel subfamily KQT member 5 (KCNQ5) as a candidate factor related to incidence of irinotecan-induced diarrhea. Irinotecan 144-154 potassium voltage-gated channel subfamily Q member 5 Homo sapiens 35-89 25127363-6 2014 We identified the SNP rs9351963 in potassium voltage-gated channel subfamily KQT member 5 (KCNQ5) as a candidate factor related to incidence of irinotecan-induced diarrhea. Irinotecan 144-154 potassium voltage-gated channel subfamily Q member 5 Homo sapiens 91-96 25127363-10 2014 These results suggest that rs9351963 in KCNQ5 is a possible predictive factor of incidence of diarrhea in cancer patients treated with irinotecan chemotherapy and for selecting chemotherapy regimens, such as irinotecan alone or a combination of irinotecan with a KCNQ5 opener. Irinotecan 135-145 potassium voltage-gated channel subfamily Q member 5 Homo sapiens 40-45 25127363-10 2014 These results suggest that rs9351963 in KCNQ5 is a possible predictive factor of incidence of diarrhea in cancer patients treated with irinotecan chemotherapy and for selecting chemotherapy regimens, such as irinotecan alone or a combination of irinotecan with a KCNQ5 opener. Irinotecan 135-145 potassium voltage-gated channel subfamily Q member 5 Homo sapiens 263-268 25127363-10 2014 These results suggest that rs9351963 in KCNQ5 is a possible predictive factor of incidence of diarrhea in cancer patients treated with irinotecan chemotherapy and for selecting chemotherapy regimens, such as irinotecan alone or a combination of irinotecan with a KCNQ5 opener. Irinotecan 208-218 potassium voltage-gated channel subfamily Q member 5 Homo sapiens 40-45 25127363-10 2014 These results suggest that rs9351963 in KCNQ5 is a possible predictive factor of incidence of diarrhea in cancer patients treated with irinotecan chemotherapy and for selecting chemotherapy regimens, such as irinotecan alone or a combination of irinotecan with a KCNQ5 opener. Irinotecan 208-218 potassium voltage-gated channel subfamily Q member 5 Homo sapiens 40-45 25110412-3 2014 Resistance to cisplatin, irinotecan and 5-fluorouracil chemotherapy has been shown to involve mitogen-activated protein kinase (MAPK) signaling and recent studies identified p38alpha MAPK as a mediator of resistance to various agents in CRC patients. Irinotecan 25-35 mitogen-activated protein kinase 14 Homo sapiens 174-182 24958824-0 2014 Dose-finding and pharmacokinetic study to optimize the dosing of irinotecan according to the UGT1A1 genotype of patients with cancer. Irinotecan 65-75 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-99 24958824-1 2014 PURPOSE: The risk of severe neutropenia from treatment with irinotecan is related in part to UGT1A1*28, a variant that reduces the elimination of SN-38, the active metabolite of irinotecan. Irinotecan 60-70 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-99 24958824-1 2014 PURPOSE: The risk of severe neutropenia from treatment with irinotecan is related in part to UGT1A1*28, a variant that reduces the elimination of SN-38, the active metabolite of irinotecan. Irinotecan 146-151 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-99 24958824-1 2014 PURPOSE: The risk of severe neutropenia from treatment with irinotecan is related in part to UGT1A1*28, a variant that reduces the elimination of SN-38, the active metabolite of irinotecan. Irinotecan 178-188 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-99 24958824-14 2014 CONCLUSION: The UGT1A1*28 genotype can be used to individualize dosing of irinotecan. Irinotecan 74-84 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 16-22 24968890-12 2014 CFZ significantly inhibited the growth of SW620 cells, and had synergistic inhibitory effects with CPT-11 on survival and colony formation; possibly by inhibition of NF-kappaB activation, MEK/ERK and PI3K/AKT pathway factor dephosphorylation and survivin downregulation. Irinotecan 99-105 AKT serine/threonine kinase 1 Homo sapiens 205-208 24968890-13 2014 Co-administration of CFZ and CPT-11 induced G2/M arrest, increased p21WAF1/CIP, and decreased mutant p53 and cdc25c expression. Irinotecan 29-35 tumor protein p53 Homo sapiens 101-104 24968890-13 2014 Co-administration of CFZ and CPT-11 induced G2/M arrest, increased p21WAF1/CIP, and decreased mutant p53 and cdc25c expression. Irinotecan 29-35 cell division cycle 25C Homo sapiens 109-115 24424522-2 2014 Here we show that interleukin-33 (IL-33) mediates the severe intestinal mucositis in mice treated with irinotecan (CPT-11), a commonly used cancer chemotherapeutic agent. Irinotecan 103-113 interleukin 33 Mus musculus 18-32 25054039-0 2014 Clinical usefulness of testing for UDP glucuronosyltransferase 1 family, polypeptide A1 polymorphism prior to the inititation of irinotecan-based chemotherapy. Irinotecan 129-139 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 35-87 25054039-1 2014 An association between UDP glucuronosyltransferase 1 family, polypeptide A1 (UGT1A1) polymorphisms and irinotecan-induced neutropenia has been previously reported. Irinotecan 103-113 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 23-75 25054039-1 2014 An association between UDP glucuronosyltransferase 1 family, polypeptide A1 (UGT1A1) polymorphisms and irinotecan-induced neutropenia has been previously reported. Irinotecan 103-113 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 77-83 23821377-0 2014 EGFR ligands as pharmacodynamic biomarkers in metastatic colorectal cancer patients treated with cetuximab and irinotecan. Irinotecan 111-121 epidermal growth factor receptor Homo sapiens 0-4 23821377-1 2014 This study was conducted to describe the modulation of plasma epidermal growth factor receptor (EGFR) ligands in EGFR-positive metastatic colorectal cancer (mCRC) patients during treatment with cetuximab and irinotecan and to explore the clinical implication of plasma levels" variations as potential biomarkers of benefit. Irinotecan 208-218 epidermal growth factor receptor Homo sapiens 62-94 23821377-1 2014 This study was conducted to describe the modulation of plasma epidermal growth factor receptor (EGFR) ligands in EGFR-positive metastatic colorectal cancer (mCRC) patients during treatment with cetuximab and irinotecan and to explore the clinical implication of plasma levels" variations as potential biomarkers of benefit. Irinotecan 208-218 epidermal growth factor receptor Homo sapiens 96-100 23821377-1 2014 This study was conducted to describe the modulation of plasma epidermal growth factor receptor (EGFR) ligands in EGFR-positive metastatic colorectal cancer (mCRC) patients during treatment with cetuximab and irinotecan and to explore the clinical implication of plasma levels" variations as potential biomarkers of benefit. Irinotecan 208-218 epidermal growth factor receptor Homo sapiens 113-117 23821377-8 2014 This hypothesis-generating study shows that EGFR ligands are significantly modulated by cetuximab plus irinotecan according to KRAS and BRAF mutational status, and they warrant further investigation as pharmacodynamic markers of resistance to anti-EGFRs. Irinotecan 103-113 epidermal growth factor receptor Homo sapiens 44-48 23821377-8 2014 This hypothesis-generating study shows that EGFR ligands are significantly modulated by cetuximab plus irinotecan according to KRAS and BRAF mutational status, and they warrant further investigation as pharmacodynamic markers of resistance to anti-EGFRs. Irinotecan 103-113 KRAS proto-oncogene, GTPase Homo sapiens 127-131 23821377-8 2014 This hypothesis-generating study shows that EGFR ligands are significantly modulated by cetuximab plus irinotecan according to KRAS and BRAF mutational status, and they warrant further investigation as pharmacodynamic markers of resistance to anti-EGFRs. Irinotecan 103-113 B-Raf proto-oncogene, serine/threonine kinase Homo sapiens 136-140 25083297-0 2014 Breast cancer resistance protein (BCRP) and excision repair cross complement-1 (ERCC1) expression in esophageal cancers and response to cisplatin and irinotecan based chemotherapy. Irinotecan 150-160 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 0-32 25083297-0 2014 Breast cancer resistance protein (BCRP) and excision repair cross complement-1 (ERCC1) expression in esophageal cancers and response to cisplatin and irinotecan based chemotherapy. Irinotecan 150-160 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 34-38 25083297-0 2014 Breast cancer resistance protein (BCRP) and excision repair cross complement-1 (ERCC1) expression in esophageal cancers and response to cisplatin and irinotecan based chemotherapy. Irinotecan 150-160 ERCC excision repair 1, endonuclease non-catalytic subunit Homo sapiens 44-78 25083297-0 2014 Breast cancer resistance protein (BCRP) and excision repair cross complement-1 (ERCC1) expression in esophageal cancers and response to cisplatin and irinotecan based chemotherapy. Irinotecan 150-160 ERCC excision repair 1, endonuclease non-catalytic subunit Homo sapiens 80-85 25083297-5 2014 We examined the expression of BCRP and ERCC1 in patients with esophageal cancer and correlated it with survival in patients receiving irinotecan and cisplatin based chemotherapy. Irinotecan 134-144 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 30-34 24513691-0 2014 Prospective study of EGFR intron 1 (CA)n repeats variants as predictors of benefit from cetuximab and irinotecan in chemo-refractory metastatic colorectal cancer (mCRC) patients. Irinotecan 102-112 epidermal growth factor receptor Homo sapiens 21-25 24824514-9 2014 Moreover, the ability of doxorubicin, SN-38 and CDDP to induce proapoptotic signals was weaker in Rho cells, as evidenced by survivin upregulation and reductions in Bax/Bcl-2 expression ratios. Irinotecan 38-43 BCL2 associated X, apoptosis regulator Homo sapiens 165-168 24952429-0 2014 Inflammasome activation is reactive oxygen species dependent and mediates irinotecan-induced mucositis through IL-1beta and IL-18 in mice. Irinotecan 74-84 interleukin 1 beta Mus musculus 111-119 24952429-0 2014 Inflammasome activation is reactive oxygen species dependent and mediates irinotecan-induced mucositis through IL-1beta and IL-18 in mice. Irinotecan 74-84 interleukin 18 Mus musculus 124-129 24952429-8 2014 Irinotecan treatment resulted in increased IL-1beta and IL-18 production in ileum and NOX-2-dependent oxidative stress. Irinotecan 0-10 interleukin 1 beta Mus musculus 43-51 24952429-8 2014 Irinotecan treatment resulted in increased IL-1beta and IL-18 production in ileum and NOX-2-dependent oxidative stress. Irinotecan 0-10 interleukin 18 Mus musculus 56-61 24952429-8 2014 Irinotecan treatment resulted in increased IL-1beta and IL-18 production in ileum and NOX-2-dependent oxidative stress. Irinotecan 0-10 cytochrome b-245, beta polypeptide Mus musculus 86-91 24952429-13 2014 NOX-2-derived oxidative stress mediates inflammasome activation and inflammasome-dependent production of IL-1beta and IL-18, which mediate tissue injury during irinotecan-induced mucositis in mice. Irinotecan 160-170 cytochrome b-245, beta polypeptide Mus musculus 0-5 24952429-13 2014 NOX-2-derived oxidative stress mediates inflammasome activation and inflammasome-dependent production of IL-1beta and IL-18, which mediate tissue injury during irinotecan-induced mucositis in mice. Irinotecan 160-170 interleukin 1 beta Mus musculus 105-113 24952429-13 2014 NOX-2-derived oxidative stress mediates inflammasome activation and inflammasome-dependent production of IL-1beta and IL-18, which mediate tissue injury during irinotecan-induced mucositis in mice. Irinotecan 160-170 interleukin 18 Mus musculus 118-123 24780296-2 2014 Bacteria beta-glucuronidase (GUS) enzymes in intestines convert the nontoxic metabolite of CPT-11, SN-38G, to toxic SN-38, and finally lead to damage of intestinal epithelial cells and diarrhea. Irinotecan 91-97 glucuronidase, beta Mus musculus 9-27 24780296-2 2014 Bacteria beta-glucuronidase (GUS) enzymes in intestines convert the nontoxic metabolite of CPT-11, SN-38G, to toxic SN-38, and finally lead to damage of intestinal epithelial cells and diarrhea. Irinotecan 91-97 glucuronidase, beta Mus musculus 29-32 24780296-2 2014 Bacteria beta-glucuronidase (GUS) enzymes in intestines convert the nontoxic metabolite of CPT-11, SN-38G, to toxic SN-38, and finally lead to damage of intestinal epithelial cells and diarrhea. Irinotecan 99-104 glucuronidase, beta Mus musculus 9-27 24780296-2 2014 Bacteria beta-glucuronidase (GUS) enzymes in intestines convert the nontoxic metabolite of CPT-11, SN-38G, to toxic SN-38, and finally lead to damage of intestinal epithelial cells and diarrhea. Irinotecan 99-104 glucuronidase, beta Mus musculus 29-32 24780296-2 2014 Bacteria beta-glucuronidase (GUS) enzymes in intestines convert the nontoxic metabolite of CPT-11, SN-38G, to toxic SN-38, and finally lead to damage of intestinal epithelial cells and diarrhea. Irinotecan 116-121 glucuronidase, beta Mus musculus 9-27 24780296-2 2014 Bacteria beta-glucuronidase (GUS) enzymes in intestines convert the nontoxic metabolite of CPT-11, SN-38G, to toxic SN-38, and finally lead to damage of intestinal epithelial cells and diarrhea. Irinotecan 116-121 glucuronidase, beta Mus musculus 29-32 24940606-0 2014 miR-345 in metastatic colorectal cancer: a non-invasive biomarker for clinical outcome in non-KRAS mutant patients treated with 3rd line cetuximab and irinotecan. Irinotecan 151-161 microRNA 345 Homo sapiens 0-7 24940606-9 2014 In addition, high miR-345 expression was associated with lack of response to treatment with cetuximab and irinotecan. Irinotecan 106-116 microRNA 345 Homo sapiens 18-25 24462762-3 2014 The study group received irinotecan dose escalation based on UGT1A1 genotyping whereas the control group did not. Irinotecan 25-35 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 61-67 24462762-8 2014 Patients with mCRC undergoing UGT1A1 genotyping may receive escalated doses of irinotecan to obtain a better clinical response/outcome with comparable toxicities. Irinotecan 79-89 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 30-36 25056111-0 2014 Reprogramming ovarian and breast cancer cells into non-cancerous cells by low-dose metformin or SN-38 through FOXO3 activation. Irinotecan 96-101 forkhead box O3 Homo sapiens 110-115 25056111-5 2014 Low-dose metformin or SN-38 increases FOXO3 nuclear localization as well as the amount of DNA damage markers and downregulates the expression of a cancer-stemness marker CD44 and other stemness markers, including Nanog, Oct-4, and c-Myc, in these cancer cells. Irinotecan 22-27 forkhead box O3 Homo sapiens 38-43 25056111-5 2014 Low-dose metformin or SN-38 increases FOXO3 nuclear localization as well as the amount of DNA damage markers and downregulates the expression of a cancer-stemness marker CD44 and other stemness markers, including Nanog, Oct-4, and c-Myc, in these cancer cells. Irinotecan 22-27 CD44 molecule (Indian blood group) Homo sapiens 170-174 25056111-5 2014 Low-dose metformin or SN-38 increases FOXO3 nuclear localization as well as the amount of DNA damage markers and downregulates the expression of a cancer-stemness marker CD44 and other stemness markers, including Nanog, Oct-4, and c-Myc, in these cancer cells. Irinotecan 22-27 Nanog homeobox Homo sapiens 213-218 25056111-5 2014 Low-dose metformin or SN-38 increases FOXO3 nuclear localization as well as the amount of DNA damage markers and downregulates the expression of a cancer-stemness marker CD44 and other stemness markers, including Nanog, Oct-4, and c-Myc, in these cancer cells. Irinotecan 22-27 POU class 5 homeobox 1 Homo sapiens 220-225 25056111-5 2014 Low-dose metformin or SN-38 increases FOXO3 nuclear localization as well as the amount of DNA damage markers and downregulates the expression of a cancer-stemness marker CD44 and other stemness markers, including Nanog, Oct-4, and c-Myc, in these cancer cells. Irinotecan 22-27 MYC proto-oncogene, bHLH transcription factor Homo sapiens 231-236 25056111-8 2014 These results suggest that low-dose metformin or SN-38 may reprogram these cancer cells into non-cancerous cells in a FOXO3-dependent manner, and may allow patients to overcome these cancers with minimal side effects. Irinotecan 49-54 forkhead box O3 Homo sapiens 118-123 24867782-0 2014 Irinotecan and temozolomide brain distribution: a focus on ABCB1. Irinotecan 0-10 ATP-binding cassette, sub-family B (MDR/TAP), member 1B Mus musculus 59-64 24867782-6 2014 Our results show that TMZ, CPT-11 and SN-38 are transported by ABCB1 at the BBB level with brain/plasma ratios of 1.1, 2.1 and 2.3, respectively. Irinotecan 27-33 ATP-binding cassette, sub-family B (MDR/TAP), member 1B Mus musculus 63-68 24867782-6 2014 Our results show that TMZ, CPT-11 and SN-38 are transported by ABCB1 at the BBB level with brain/plasma ratios of 1.1, 2.1 and 2.3, respectively. Irinotecan 38-43 ATP-binding cassette, sub-family B (MDR/TAP), member 1B Mus musculus 63-68 24727322-9 2014 Everolimus also markedly inhibited the hydrolysis of irinotecan and p-nitrophenyl acetate by mouse plasma carboxylesterase and recombinant human CES2, respectively. Irinotecan 53-63 carboxylesterase 2 Homo sapiens 145-149 24824514-9 2014 Moreover, the ability of doxorubicin, SN-38 and CDDP to induce proapoptotic signals was weaker in Rho cells, as evidenced by survivin upregulation and reductions in Bax/Bcl-2 expression ratios. Irinotecan 38-43 BCL2 apoptosis regulator Homo sapiens 169-174 24726739-2 2014 Our results indicate for the first time that linsitinib significantly potentiate the effect of anti-neoplastic drugs mitoxantrone (MX) and SN-38 in ABCG2-overexpressing cells; paclitaxel, docetaxel and vinblastine in ABCC10-overexpressing cells. Irinotecan 139-144 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 148-153 24977443-0 2014 [Interest of UGT1A1 genotyping within digestive cancers treatment by irinotecan]. Irinotecan 69-79 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 13-19 24977443-2 2014 Its anticancer activity results from its bioactivation into SN-38 metabolite, which is cleared through glucuronidation by the hepatic enzyme uridine diphosphate-glucuronosyltransferase 1A1 (UGT1A1). Irinotecan 60-65 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 141-188 24977443-2 2014 Its anticancer activity results from its bioactivation into SN-38 metabolite, which is cleared through glucuronidation by the hepatic enzyme uridine diphosphate-glucuronosyltransferase 1A1 (UGT1A1). Irinotecan 60-65 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 190-196 24977443-9 2014 The existence of a French laboratories network performing this test in clinical routine makes it possible to generalize UGT1A1 deficiency screening, so as to guarantee equal access to safe treatment and optimized irinorecan-based therapy for the many patients receiving irinotecan-based therapy in advanced colorectal cancer. Irinotecan 270-280 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 120-126 24478107-5 2014 RS4 consumption compared with CF resulted in 7.2% (p = 0.002) lower mean total cholesterol, 5.5% (p = 0.04) lower non-HDL, and a 12.8% (p < 0.001) lower HDL cholesterol in the With-MetS group. Irinotecan 0-3 ETS variant transcription factor 3 Homo sapiens 179-188 24468885-2 2014 Therefore, we designed a study using simvastatin/cetuximab/irinotecan for KRAS mutant CRC patients who are refractory to irinotecan and oxaliplatin-based chemotherapy. Irinotecan 59-69 KRAS proto-oncogene, GTPase Homo sapiens 74-78 24468885-2 2014 Therefore, we designed a study using simvastatin/cetuximab/irinotecan for KRAS mutant CRC patients who are refractory to irinotecan and oxaliplatin-based chemotherapy. Irinotecan 121-131 KRAS proto-oncogene, GTPase Homo sapiens 74-78 24468885-13 2014 CONCLUSION: The simvastatin/cetuximab/irinotecan regimen showed promising efficacy and safety in KRAS mutant CRC patients who failed irinotecan and oxaliplatin-based chemotherapy. Irinotecan 38-48 KRAS proto-oncogene, GTPase Homo sapiens 97-101 24627085-9 2014 The veliparib and CPT-11 combination can be further explored as a treatment of metastatic castration-resistant prostate cancers that have frequent p53 mutations and enriched genomic instability. Irinotecan 18-24 tumor protein p53 L homeolog Xenopus laevis 147-150 24453052-1 2014 BACKGROUND AND OBJECTIVES: Uridine-diphosphate glucuronosyltransferase 1A (UGT1A) is a key enzyme involved in irinotecan metabolism, and polymorphisms in the UGT1A gene are associated with irinotecan-induced toxicity. Irinotecan 110-120 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 27-73 24453052-1 2014 BACKGROUND AND OBJECTIVES: Uridine-diphosphate glucuronosyltransferase 1A (UGT1A) is a key enzyme involved in irinotecan metabolism, and polymorphisms in the UGT1A gene are associated with irinotecan-induced toxicity. Irinotecan 110-120 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 75-80 24453052-1 2014 BACKGROUND AND OBJECTIVES: Uridine-diphosphate glucuronosyltransferase 1A (UGT1A) is a key enzyme involved in irinotecan metabolism, and polymorphisms in the UGT1A gene are associated with irinotecan-induced toxicity. Irinotecan 110-120 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 158-163 24453052-1 2014 BACKGROUND AND OBJECTIVES: Uridine-diphosphate glucuronosyltransferase 1A (UGT1A) is a key enzyme involved in irinotecan metabolism, and polymorphisms in the UGT1A gene are associated with irinotecan-induced toxicity. Irinotecan 189-199 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 27-73 24453052-1 2014 BACKGROUND AND OBJECTIVES: Uridine-diphosphate glucuronosyltransferase 1A (UGT1A) is a key enzyme involved in irinotecan metabolism, and polymorphisms in the UGT1A gene are associated with irinotecan-induced toxicity. Irinotecan 189-199 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 75-80 24453052-1 2014 BACKGROUND AND OBJECTIVES: Uridine-diphosphate glucuronosyltransferase 1A (UGT1A) is a key enzyme involved in irinotecan metabolism, and polymorphisms in the UGT1A gene are associated with irinotecan-induced toxicity. Irinotecan 189-199 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 158-163 24453052-10 2014 A comprehensive study of the influence of UGT1A1 polymorphisms on the risk of irinotecan-induced toxicity is necessary for optimal use of irinotecan treatment. Irinotecan 78-88 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 42-48 24453052-10 2014 A comprehensive study of the influence of UGT1A1 polymorphisms on the risk of irinotecan-induced toxicity is necessary for optimal use of irinotecan treatment. Irinotecan 138-148 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 42-48 23959273-0 2014 Intergenic polymorphisms in the amphiregulin gene region as biomarkers in metastatic colorectal cancer patients treated with anti-EGFR plus irinotecan. Irinotecan 140-150 amphiregulin Homo sapiens 32-44 24932285-0 2014 Association between the low-dose irinotecan regimen-induced occurrence of grade 4 neutropenia and genetic variants of UGT1A1 in patients with gynecological cancers. Irinotecan 33-43 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 118-124 24932285-1 2014 The occurrence of severe neutropenia during treatment with irinotecan (CPT-11) is associated with the *6 and *28 alleles of uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1). Irinotecan 59-69 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 124-171 24932285-1 2014 The occurrence of severe neutropenia during treatment with irinotecan (CPT-11) is associated with the *6 and *28 alleles of uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1). Irinotecan 59-69 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 173-179 24932285-1 2014 The occurrence of severe neutropenia during treatment with irinotecan (CPT-11) is associated with the *6 and *28 alleles of uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1). Irinotecan 71-77 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 124-171 24932285-1 2014 The occurrence of severe neutropenia during treatment with irinotecan (CPT-11) is associated with the *6 and *28 alleles of uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1). Irinotecan 71-77 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 173-179 24932285-3 2014 There are also no studies regarding the association between the 421C>A mutation in ATP-binding cassette sub-family G member 2 (ABCG2) and the occurrence of severe neutropenia in CPT-11-treated patients with gynecological cancers. Irinotecan 181-187 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 86-128 24932285-3 2014 There are also no studies regarding the association between the 421C>A mutation in ATP-binding cassette sub-family G member 2 (ABCG2) and the occurrence of severe neutropenia in CPT-11-treated patients with gynecological cancers. Irinotecan 181-187 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 130-135 24932285-6 2014 The association between the absolute neutrophil count (ANC) nadir values, the total dose of CPT-11 and the genotypes of UGT1A1 or ABCG2 was studied. Irinotecan 92-98 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 120-126 24932285-12 2014 In conclusion, the UGT1A1*6/*28 and *6/*6 genotypes were found to be associated with the occurrence of severe neutropenia in the low-dose CPT-11 regimen for gynecological cancers. Irinotecan 138-144 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 19-25 24932285-13 2014 This finding indicates that the determination of UGT1A1 variants may be as useful in CPT-11 chemotherapy for gynecological conditions as it is in colorectal and lung cancer patients treated with this drug. Irinotecan 85-91 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 49-55 25141892-0 2014 UGT1A1*28 polymorphisms: a potential pharmacological biomarker of irinotecan-based chemotherapies in colorectal cancer. Irinotecan 66-76 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 23959273-8 2014 Our findings support that intergenic polymorphisms in the AREG gene region might help to identify colorectal cancer patients that will benefit from irinotecan plus anti-EGFR therapy. Irinotecan 148-158 amphiregulin Homo sapiens 58-62 24959385-14 2014 Furthermore, both irinotecan and topotecan are the efflux pump ABCG2 substrates; cancer cells with high expression of ABCG2 showed strong irinotecan and topotecan resistance. Irinotecan 18-28 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 63-68 24959385-14 2014 Furthermore, both irinotecan and topotecan are the efflux pump ABCG2 substrates; cancer cells with high expression of ABCG2 showed strong irinotecan and topotecan resistance. Irinotecan 18-28 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 118-123 24959385-14 2014 Furthermore, both irinotecan and topotecan are the efflux pump ABCG2 substrates; cancer cells with high expression of ABCG2 showed strong irinotecan and topotecan resistance. Irinotecan 138-148 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 63-68 24959385-14 2014 Furthermore, both irinotecan and topotecan are the efflux pump ABCG2 substrates; cancer cells with high expression of ABCG2 showed strong irinotecan and topotecan resistance. Irinotecan 138-148 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 118-123 24798549-11 2014 Reovirus and irinotecan combination therapy is synergistic, p21 mediated, and represents a novel potential treatment for patients with CRC. Irinotecan 13-23 H3 histone pseudogene 16 Homo sapiens 60-63 24611457-10 2014 Thus, MTHFR genetic polymorphisms may be screened to predict the clinical responses to irinotecan-based chemotherapy in CRC patients. Irinotecan 87-97 methylenetetrahydrofolate reductase Homo sapiens 6-11 24428790-0 2014 Targeted inhibition of IL-18 attenuates irinotecan-induced intestinal mucositis in mice. Irinotecan 40-50 interleukin 18 Mus musculus 23-28 24428790-4 2014 Therefore, we have investigated the role of IL-18 in the pathogenesis of irinotecan-induced intestinal mucositis. Irinotecan 73-83 interleukin 18 Mus musculus 44-49 24428790-9 2014 Additionally, irinotecan treatment increased MPO and iNOS activity, iNOS immunostaining and IL-18 expression in WT mice compared with saline treatment. Irinotecan 14-24 myeloperoxidase Mus musculus 45-48 24428790-9 2014 Additionally, irinotecan treatment increased MPO and iNOS activity, iNOS immunostaining and IL-18 expression in WT mice compared with saline treatment. Irinotecan 14-24 nitric oxide synthase 2, inducible Mus musculus 53-57 24428790-9 2014 Additionally, irinotecan treatment increased MPO and iNOS activity, iNOS immunostaining and IL-18 expression in WT mice compared with saline treatment. Irinotecan 14-24 nitric oxide synthase 2, inducible Mus musculus 68-72 24428790-9 2014 Additionally, irinotecan treatment increased MPO and iNOS activity, iNOS immunostaining and IL-18 expression in WT mice compared with saline treatment. Irinotecan 14-24 interleukin 18 Mus musculus 92-97 24428790-13 2014 Furthermore, the Survival of irinotecan-treated mice was increased and iNOS immunoexpression and IL-18 production prevented in IL-18 knockout mice. Irinotecan 29-39 interleukin 18 Mus musculus 127-132 24428790-14 2014 CONCLUSIONS AND IMPLICATIONS: Targeting IL-18 function may be a promising therapeutic approach to decreasing the severity of intestinal mucositis during irinotecan treatment regimens. Irinotecan 153-163 interleukin 18 Mus musculus 40-45 24834123-1 2014 Uridine glucuronosyltransferase (UGT) gene polymorphisms have been linked to irinotecan toxicity. Irinotecan 77-87 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 0-31 24834123-1 2014 Uridine glucuronosyltransferase (UGT) gene polymorphisms have been linked to irinotecan toxicity. Irinotecan 77-87 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 33-36 24834123-2 2014 Our purpose was to study the association between UGT1A1*28, UGT1A7*2, and UGT1A7*3 polymorphisms and irinotecan toxicity in Greek patients receiving low-dose weekly irinotecan. Irinotecan 101-111 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 49-55 24834123-2 2014 Our purpose was to study the association between UGT1A1*28, UGT1A7*2, and UGT1A7*3 polymorphisms and irinotecan toxicity in Greek patients receiving low-dose weekly irinotecan. Irinotecan 101-111 UDP glucuronosyltransferase family 1 member A7 Homo sapiens 74-80 24834123-8 2014 To conclude, UGT polymorphisms play a role in the toxicity of irinotecan, even if the drug is administered in low doses. Irinotecan 62-72 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 13-16 24611457-0 2014 Effects of MTHFR genetic polymorphisms on toxicity and clinical response of irinotecan-based chemotherapy in patients with colorectal cancer. Irinotecan 76-86 methylenetetrahydrofolate reductase Homo sapiens 11-16 24611457-1 2014 AIMS: This meta-analysis aims to evaluate the effects of common polymorphisms in the methylenetetrahydrofolate reductase (MTHFR) gene on the toxicity and clinical responses of irinotecan-based chemotherapy in patients with colorectal cancer (CRC). Irinotecan 176-186 methylenetetrahydrofolate reductase Homo sapiens 85-120 24611457-1 2014 AIMS: This meta-analysis aims to evaluate the effects of common polymorphisms in the methylenetetrahydrofolate reductase (MTHFR) gene on the toxicity and clinical responses of irinotecan-based chemotherapy in patients with colorectal cancer (CRC). Irinotecan 176-186 methylenetetrahydrofolate reductase Homo sapiens 122-127 24611457-7 2014 RESULTS: The results from our meta-analysis suggested that MTHFR genetic polymorphisms might significantly decrease the rate of grade 3/4 toxicity of irinotecan-based chemotherapy in CRC patients (OR=0.53, 95% CI: 0.32-0.89, p=0.015). Irinotecan 150-160 methylenetetrahydrofolate reductase Homo sapiens 59-64 24611457-8 2014 Furthermore, we also demonstrated that MTHFR genetic polymorphisms strongly correlated with good clinical responses (complete response+partial response) to irinotecan-based chemotherapy in CRC patients (OR=1.47, 95% CI: 1.05-2.04, p=0.024). Irinotecan 156-166 methylenetetrahydrofolate reductase Homo sapiens 39-44 24611457-9 2014 CONCLUSIONS: Our findings provide empirical evidence that MTHFR genetic polymorphisms may decrease the toxicity of irinotecan-based chemotherapy and increase the clinical benefits for CRC patients. Irinotecan 115-125 methylenetetrahydrofolate reductase Homo sapiens 58-63 24382596-1 2014 Previous reports of the influence of UGT1A1 gene polymorphisms on the pharmacokinetics of irinotecan metabolism have not assessed Asian patients treated with FOLFIRI plus bevacizumab for advanced and recurrent colorectal cancer. Irinotecan 90-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 37-43 24692733-0 2014 Multicenter phase II study of second-line cetuximab plus folinic acid/5-fluorouracil/irinotecan (FOLFIRI) in KRAS wild-type metastatic colorectal cancer: the FLIER study. Irinotecan 85-95 KRAS proto-oncogene, GTPase Homo sapiens 109-113 24683007-1 2014 Weekly cetuximab plus irinotecan-based regiments are standard first- and second-line chemotherapy for patients with KRAS wild-type metastatic colorectal cancer (mCRC). Irinotecan 22-32 KRAS proto-oncogene, GTPase Homo sapiens 116-120 24249672-2 2014 A phase I trial of the mTOR inhibitor temsirolimus (TEM) with irinotecan (IRN) and temozolomide (TMZ) was conducted in children with recurrent/refractory solid tumors, including central nervous system (CNS) tumors. Irinotecan 62-72 mechanistic target of rapamycin kinase Homo sapiens 23-27 24249672-2 2014 A phase I trial of the mTOR inhibitor temsirolimus (TEM) with irinotecan (IRN) and temozolomide (TMZ) was conducted in children with recurrent/refractory solid tumors, including central nervous system (CNS) tumors. Irinotecan 74-77 mechanistic target of rapamycin kinase Homo sapiens 23-27 24764659-5 2014 The CRYSTAL study showed that adding cetuximab to FOLFIRI (regimen of irinotecan, infusional fluorouracil and leucovorin) significantly improved results in the first-line treatment of KRAS wild-type mCRC. Irinotecan 70-80 KRAS proto-oncogene, GTPase Homo sapiens 184-188 24699684-1 2014 Two major forms of human carboxylesterase (CES), CES1A and CES2, dominate the pharmacokinetics of most prodrugs such as imidapril and irinotecan (CPT-11). Irinotecan 134-144 carboxylesterase 2 Homo sapiens 59-63 24699684-1 2014 Two major forms of human carboxylesterase (CES), CES1A and CES2, dominate the pharmacokinetics of most prodrugs such as imidapril and irinotecan (CPT-11). Irinotecan 146-152 carboxylesterase 2 Homo sapiens 59-63 24577941-0 2014 The use of Olaparib (AZD2281) potentiates SN-38 cytotoxicity in colon cancer cells by indirect inhibition of Rad51-mediated repair of DNA double-strand breaks. Irinotecan 42-47 RAD51 recombinase Mus musculus 109-114 24577941-5 2014 Furthermore, olaparib potentiated S-phase-specific double-strand DNA breaks (DSB) induced by SN-38, which is followed by Rad51 recruitment. Irinotecan 93-98 RAD51 recombinase Mus musculus 121-126 24577941-6 2014 siRNA-mediated knockdown of Rad51, but not Mre11 or Rad50, increased the sensitivity to olaparib and/or SN-38 treatment in colon cancer cells. Irinotecan 104-109 RAD51 recombinase Mus musculus 28-33 24772300-6 2014 The mPFS of the wild-type and mutated KRAS subgroups that had received irinotecan-based treatments was 7.7 and 9.7 months, respectively (P= 0.43). Irinotecan 71-81 KRAS proto-oncogene, GTPase Homo sapiens 38-42 24781822-3 2014 SLC6A6 knockdown (KD) attenuated cell survival and was accompanied by enhanced drug sensitivity to 5-fluorouracil (5-FU), doxycycline (DOX) and SN-38. Irinotecan 144-149 solute carrier family 6 member 6 Homo sapiens 0-6 24781822-6 2014 Additionally, SLC6A6-siRNA treatment enhanced the cytotoxic effects of all 3 drugs, whereas the efficacy of ABCG2-siRNA treatment was limited to its 2 substrate drugs, DOX and SN-38. Irinotecan 176-181 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 108-113 24692733-1 2014 BACKGROUND: This study was the first multicenter phase II study of cetuximab plus folinic acid/5-fluorouracil/irinotecan (FOLFIRI) in KRAS wild-type mCRC as a second-line treatment in Japan including BRAF and PIK3CA genotyping. Irinotecan 110-120 KRAS proto-oncogene, GTPase Homo sapiens 134-138 24287113-5 2014 The highest killing efficiency was observed after co-incubation of the cells with irinotecan and vitamin A (10muM), or vitamin E (25muM), respectively. Irinotecan 82-92 latexin Homo sapiens 110-113 24519753-0 2014 Associations between UGT1A1*6 or UGT1A1*6/*28 polymorphisms and irinotecan-induced neutropenia in Asian cancer patients. Irinotecan 64-74 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-39 24519753-2 2014 Many studies have demonstrated that patients bearing UGT1A1*28 have a higher risk of severe neutropenia on toxicity of irinotecan. Irinotecan 119-129 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 53-59 24519753-4 2014 Some researches reported that UGT1A1*28 and/or UGT1A1*6 could predict irinotecan-induced toxicities in Asian populations, but controversial conclusions still remained. Irinotecan 70-80 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 30-36 24519753-4 2014 Some researches reported that UGT1A1*28 and/or UGT1A1*6 could predict irinotecan-induced toxicities in Asian populations, but controversial conclusions still remained. Irinotecan 70-80 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 47-53 24519753-5 2014 This study aims to investigate the association between UGT1A1 gene polymorphisms *6, *6/*28 and irinotecan-related neutropenia in Asian cancer patients receiving irinotecan regimen chemotherapy. Irinotecan 96-106 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 55-61 24519753-5 2014 This study aims to investigate the association between UGT1A1 gene polymorphisms *6, *6/*28 and irinotecan-related neutropenia in Asian cancer patients receiving irinotecan regimen chemotherapy. Irinotecan 162-172 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 55-61 24519753-6 2014 EXPERIMENTAL DESIGN: Meta-analyses were done to assess the relationship between UGT1A1*6 or UGT1A1*6/*28 and irinotecan-induced neutropenia. Irinotecan 109-119 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 80-86 24519753-6 2014 EXPERIMENTAL DESIGN: Meta-analyses were done to assess the relationship between UGT1A1*6 or UGT1A1*6/*28 and irinotecan-induced neutropenia. Irinotecan 109-119 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 92-98 24519753-9 2014 CONCLUSIONS: In conclusion, the UGT1A1*6 and UGT1A1*6/*28 genotypes were associated with an increased risk of irinotecan-induced neutropenia in Asian cancer patients. Irinotecan 110-120 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 32-38 24519753-9 2014 CONCLUSIONS: In conclusion, the UGT1A1*6 and UGT1A1*6/*28 genotypes were associated with an increased risk of irinotecan-induced neutropenia in Asian cancer patients. Irinotecan 110-120 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 45-51 24242862-2 2014 Camptothecin analogues, including SN-38, have been shown to reduce the expression and transcriptional activity of HIF-1alpha in preclinical models. Irinotecan 34-39 hypoxia inducible factor 1 subunit alpha Homo sapiens 114-124 24242862-3 2014 We hypothesized that co-administration of pegylated SN-38 (EZN-2208) may offset the induction of HIF-1alpha following bevacizumab treatment, resulting in synergistic antitumor effects. Irinotecan 52-57 hypoxia inducible factor 1 subunit alpha Homo sapiens 97-107 24897286-0 2014 Association analysis of UGT1A genotype and haplotype with SN-38 glucuronidation in human livers. Irinotecan 58-63 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 24-29 24897286-1 2014 AIM: 7-ethyl-10-hydroxycamptothecin (SN-38), the active metabolite of irinotecan, is mainly eliminated hepatically through glucuronidation by UGT1A1 and UGT1A9 enzymes. Irinotecan 5-35 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 142-148 24897286-1 2014 AIM: 7-ethyl-10-hydroxycamptothecin (SN-38), the active metabolite of irinotecan, is mainly eliminated hepatically through glucuronidation by UGT1A1 and UGT1A9 enzymes. Irinotecan 5-35 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 153-159 24897286-1 2014 AIM: 7-ethyl-10-hydroxycamptothecin (SN-38), the active metabolite of irinotecan, is mainly eliminated hepatically through glucuronidation by UGT1A1 and UGT1A9 enzymes. Irinotecan 37-42 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 142-148 24897286-1 2014 AIM: 7-ethyl-10-hydroxycamptothecin (SN-38), the active metabolite of irinotecan, is mainly eliminated hepatically through glucuronidation by UGT1A1 and UGT1A9 enzymes. Irinotecan 37-42 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 153-159 24897286-1 2014 AIM: 7-ethyl-10-hydroxycamptothecin (SN-38), the active metabolite of irinotecan, is mainly eliminated hepatically through glucuronidation by UGT1A1 and UGT1A9 enzymes. Irinotecan 70-80 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 142-148 24897286-1 2014 AIM: 7-ethyl-10-hydroxycamptothecin (SN-38), the active metabolite of irinotecan, is mainly eliminated hepatically through glucuronidation by UGT1A1 and UGT1A9 enzymes. Irinotecan 70-80 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 153-159 24897286-2 2014 This study comprehensively investigates the effects of UGT1A1 and UGT1A9 genetic polymorphism on SN-38 glucuronidation activity. Irinotecan 97-102 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 55-61 24897286-2 2014 This study comprehensively investigates the effects of UGT1A1 and UGT1A9 genetic polymorphism on SN-38 glucuronidation activity. Irinotecan 97-102 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 66-72 24897286-5 2014 The number of UGT1A1 reduced function alleles was associated with decreased SN-38G formation rates and UGT1A protein levels. Irinotecan 76-81 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 14-20 24897286-5 2014 The number of UGT1A1 reduced function alleles was associated with decreased SN-38G formation rates and UGT1A protein levels. Irinotecan 76-81 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 14-19 24897286-6 2014 UGT1A9 I399C>T and UGT1A9*1b, which were highly linked, were associated with increased SN-38 glucuronidation activity and UGT1A protein levels. Irinotecan 90-95 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 0-6 24897286-6 2014 UGT1A9 I399C>T and UGT1A9*1b, which were highly linked, were associated with increased SN-38 glucuronidation activity and UGT1A protein levels. Irinotecan 90-95 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 22-28 24897286-6 2014 UGT1A9 I399C>T and UGT1A9*1b, which were highly linked, were associated with increased SN-38 glucuronidation activity and UGT1A protein levels. Irinotecan 90-95 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 0-5 24897286-8 2014 CONCLUSION: UGT1A1 genetic polymorphisms have a more important function in human liver SN-38 glucuronidation activity than UGT1A9. Irinotecan 87-92 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-18 24407191-0 2014 Sorafenib and irinotecan (NEXIRI) as second- or later-line treatment for patients with metastatic colorectal cancer and KRAS-mutated tumours: a multicentre Phase I/II trial. Irinotecan 14-24 KRAS proto-oncogene, GTPase Homo sapiens 120-124 24318863-5 2014 Target genes were XPD-751, GSTP-1-105, XRCC1-399 for oxaliplatin, UGT1A1 for irinotecan. Irinotecan 77-87 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 66-72 23504048-5 2014 Single and combined use of paclitaxel and SN-38 produced significant cytolethality against the cervical adenocarcinoma cell line CAC-1. Irinotecan 42-47 transmembrane protein 54 Homo sapiens 129-134 24215868-4 2014 Synergism was evident between LT-626 and cisplatin, oxaliplatin and SN-38 suggesting that PARP inhibitors in combination with DNA damaging agents may be a successful strategy for treatment of CRC. Irinotecan 68-73 poly(ADP-ribose) polymerase 1 Homo sapiens 90-94 24462359-4 2014 In addition, P123-DOPE can inhibit breast cancer resistance protein (BCPR) mediated CPT-11 efflux in drug resistant MCF-7/BCRP breast cancer cells, thus acting as a "door blocker". Irinotecan 84-90 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 35-67 24462359-4 2014 In addition, P123-DOPE can inhibit breast cancer resistance protein (BCPR) mediated CPT-11 efflux in drug resistant MCF-7/BCRP breast cancer cells, thus acting as a "door blocker". Irinotecan 84-90 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 69-73 23384250-1 2014 ATP-Binding Cassette transporters such as ABCG2 confer resistance to various anticancer drugs including irinotecan and its active metabolite, SN38. Irinotecan 104-114 ATP binding cassette subfamily G member 2 (Junior blood group) Mus musculus 42-47 23605141-1 2014 BACKGROUND: It was recently reported that genetic polymorphisms of UDP glucuronyltransferase-1 polypeptide A1 (UGT1A1), a glucuronidation enzyme, were associated with irinotecan (CPT-11) metabolism. Irinotecan 167-177 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 67-109 23605141-1 2014 BACKGROUND: It was recently reported that genetic polymorphisms of UDP glucuronyltransferase-1 polypeptide A1 (UGT1A1), a glucuronidation enzyme, were associated with irinotecan (CPT-11) metabolism. Irinotecan 167-177 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 111-117 23605141-1 2014 BACKGROUND: It was recently reported that genetic polymorphisms of UDP glucuronyltransferase-1 polypeptide A1 (UGT1A1), a glucuronidation enzyme, were associated with irinotecan (CPT-11) metabolism. Irinotecan 179-185 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 67-109 23605141-1 2014 BACKGROUND: It was recently reported that genetic polymorphisms of UDP glucuronyltransferase-1 polypeptide A1 (UGT1A1), a glucuronidation enzyme, were associated with irinotecan (CPT-11) metabolism. Irinotecan 179-185 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 111-117 23605141-2 2014 The active metabolite of CPT-11, 7-ethyl-10-hydroxycamptothecin (SN-38) was glucuronidated (SN-38G) by UGT1A1. Irinotecan 65-70 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 103-109 23605141-2 2014 The active metabolite of CPT-11, 7-ethyl-10-hydroxycamptothecin (SN-38) was glucuronidated (SN-38G) by UGT1A1. Irinotecan 92-97 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 103-109 23605141-4 2014 Several studies have suggested that the dose of CPT-11 should be decreased in patients homozygous for UGT1A1*6 or UGT1A1*28, or double heterozygotes (*6/*28). Irinotecan 48-54 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 102-108 23605141-4 2014 Several studies have suggested that the dose of CPT-11 should be decreased in patients homozygous for UGT1A1*6 or UGT1A1*28, or double heterozygotes (*6/*28). Irinotecan 48-54 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 114-120 23605141-7 2014 RESULTS: The SN-38G/SN-38 concentration ratio was lower in patients who were homozygous for UGT1A1*6, heterozygous for UGT1A1*6 or UGT1A1*28, or were double heterozygotes compared with patients with wild-type genes. Irinotecan 13-18 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 92-98 23605141-7 2014 RESULTS: The SN-38G/SN-38 concentration ratio was lower in patients who were homozygous for UGT1A1*6, heterozygous for UGT1A1*6 or UGT1A1*28, or were double heterozygotes compared with patients with wild-type genes. Irinotecan 13-18 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 119-125 23605141-7 2014 RESULTS: The SN-38G/SN-38 concentration ratio was lower in patients who were homozygous for UGT1A1*6, heterozygous for UGT1A1*6 or UGT1A1*28, or were double heterozygotes compared with patients with wild-type genes. Irinotecan 13-18 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 119-125 23605141-8 2014 The relative decreases in the SN-38G/SN-38 concentration ratio in patients homozygous for UGT1A1*6 and in double heterozygotes were greater than in patients heterozygous for UGT1A1*6 or UGT1A1*28. Irinotecan 30-35 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 90-96 23605141-8 2014 The relative decreases in the SN-38G/SN-38 concentration ratio in patients homozygous for UGT1A1*6 and in double heterozygotes were greater than in patients heterozygous for UGT1A1*6 or UGT1A1*28. Irinotecan 30-35 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 174-180 23605141-8 2014 The relative decreases in the SN-38G/SN-38 concentration ratio in patients homozygous for UGT1A1*6 and in double heterozygotes were greater than in patients heterozygous for UGT1A1*6 or UGT1A1*28. Irinotecan 30-35 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 174-180 23609856-3 2014 Beside *28, UGT1A1*6 (*6) is another important variant allele in Japanese patients because it is associated with adverse events of irinotecan, metabolized by UGT1A1. Irinotecan 131-141 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-18 23609856-3 2014 Beside *28, UGT1A1*6 (*6) is another important variant allele in Japanese patients because it is associated with adverse events of irinotecan, metabolized by UGT1A1. Irinotecan 131-141 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 158-164 23529007-0 2014 Association of UGT1A1*28 polymorphisms with irinotecan-induced toxicities in colorectal cancer: a meta-analysis in Caucasians. Irinotecan 44-54 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 15-21 23529007-1 2014 A meta-analysis in Caucasians was conducted to investigate the possible association of uridine diphosphate glucuronosyltransferase (UGT) 1A1 gene polymorphisms with irinotecan (IRI)-induced neutropenia and diarrhoea in colorectal cancer (CRC). Irinotecan 165-175 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 87-140 24066723-2 2014 Nilotinib, a tyrosine kinase inhibitor, could potently inhibit SN-38 glucuronidation mainly catalyzed by UDP-glucuronosyltransferase (UGT) 1A1. Irinotecan 63-68 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 105-142 24448639-0 2014 UGT1A1*6 polymorphisms are correlated with irinotecan-induced toxicity: a system review and meta-analysis in Asians. Irinotecan 43-53 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 24525731-6 2014 Irinotecan, another chemotherapeutic agent to induce DNA damaging used to treat patients with advanced gastric cancer that progressed on cisplatin, was found to inhibit the expression of XRCC1 effectively, and leading to an increase in the sensitivity of resistant cells to cisplatin. Irinotecan 0-10 X-ray repair cross complementing 1 Homo sapiens 187-192 24523596-2 2014 ABCG2/BCRP is referred to as a "half-type" ABC transporter, functioning as a homodimer, and transports anticancer agents such as irinotecan, 7-ethyl-10-hydroxycamptothecin (SN-38), gefitinib, imatinib, methotrexate, and mitoxantrone from cells. Irinotecan 129-139 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 6-10 24523596-2 2014 ABCG2/BCRP is referred to as a "half-type" ABC transporter, functioning as a homodimer, and transports anticancer agents such as irinotecan, 7-ethyl-10-hydroxycamptothecin (SN-38), gefitinib, imatinib, methotrexate, and mitoxantrone from cells. Irinotecan 129-139 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 0-5 24523596-2 2014 ABCG2/BCRP is referred to as a "half-type" ABC transporter, functioning as a homodimer, and transports anticancer agents such as irinotecan, 7-ethyl-10-hydroxycamptothecin (SN-38), gefitinib, imatinib, methotrexate, and mitoxantrone from cells. Irinotecan 141-171 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 0-5 24523596-2 2014 ABCG2/BCRP is referred to as a "half-type" ABC transporter, functioning as a homodimer, and transports anticancer agents such as irinotecan, 7-ethyl-10-hydroxycamptothecin (SN-38), gefitinib, imatinib, methotrexate, and mitoxantrone from cells. Irinotecan 141-171 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 6-10 24523596-2 2014 ABCG2/BCRP is referred to as a "half-type" ABC transporter, functioning as a homodimer, and transports anticancer agents such as irinotecan, 7-ethyl-10-hydroxycamptothecin (SN-38), gefitinib, imatinib, methotrexate, and mitoxantrone from cells. Irinotecan 173-178 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 0-5 24523596-2 2014 ABCG2/BCRP is referred to as a "half-type" ABC transporter, functioning as a homodimer, and transports anticancer agents such as irinotecan, 7-ethyl-10-hydroxycamptothecin (SN-38), gefitinib, imatinib, methotrexate, and mitoxantrone from cells. Irinotecan 173-178 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 6-10 24316664-9 2014 mRNA levels of claudin-1 were significantly decreased early after irinotecan in the small and large intestines. Irinotecan 66-76 claudin 1 Rattus norvegicus 15-24 24439671-0 2014 Regulation and expression of aberrant methylation on irinotecan metabolic genes CES2, UGT1A1 and GUSB in the in-vitro cultured colorectal cancer cells. Irinotecan 53-63 carboxylesterase 2 Homo sapiens 80-84 24439671-0 2014 Regulation and expression of aberrant methylation on irinotecan metabolic genes CES2, UGT1A1 and GUSB in the in-vitro cultured colorectal cancer cells. Irinotecan 53-63 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 86-92 24439671-0 2014 Regulation and expression of aberrant methylation on irinotecan metabolic genes CES2, UGT1A1 and GUSB in the in-vitro cultured colorectal cancer cells. Irinotecan 53-63 glucuronidase beta Homo sapiens 97-101 24439671-8 2014 CONCLUSION: Methylation in UGT1A1 gene expression silencing as an important mechanism; methylation could provide an effective target for methylation regulation intervening in the treatment of CPT-11. Irinotecan 192-198 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 27-33 24308720-0 2014 The association of UGT1A1*6 and UGT1A1*28 with irinotecan-induced neutropenia in Asians: a meta-analysis. Irinotecan 47-57 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 19-25 24308720-0 2014 The association of UGT1A1*6 and UGT1A1*28 with irinotecan-induced neutropenia in Asians: a meta-analysis. Irinotecan 47-57 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 32-38 24308720-1 2014 BACKGROUND: The UGT1A1*28 polymorphism is known as a biomarker of irinotecan-induced neutropenia in Caucasians. Irinotecan 66-76 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 16-22 24308720-5 2014 CONCLUSIONS: In Asians, a combination test of UGT1A1*6 and UGT1A1*28 might be a potential biomarker of irinotecan-induced neutropenia, an observation that will need additional trials for confirmation. Irinotecan 103-113 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 46-52 24308720-5 2014 CONCLUSIONS: In Asians, a combination test of UGT1A1*6 and UGT1A1*28 might be a potential biomarker of irinotecan-induced neutropenia, an observation that will need additional trials for confirmation. Irinotecan 103-113 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 59-65 24265452-10 2014 Finally, in irinotecan-induced enteritis, GLP-2 normalized epithelial barrier function in control (P < .05) but not knockout animals. Irinotecan 12-22 glucagon-like peptide 2 receptor Mus musculus 42-47 24497922-1 2014 OBJECTIVE: To explore genes of the killer-cell immunoglobulin-like receptor (KIR) and of the HLA ligand and their relationship with the outcome of metastatic colorectal cancer (mCRC) patients treated with first-line 5-fluorouracil, leucovorin, and irinotecan (FOLFIRI). Irinotecan 248-258 killer cell immunoglobulin like receptor, two Ig domains and short cytoplasmic tail 4 Homo sapiens 35-75 24194565-2 2014 Impaired hepatic clearance due to low-activity polymorphisms in human OATP1B1 may increase systemic exposure to SN-38, the active and toxic metabolite of the anticancer prodrug irinotecan. Irinotecan 112-117 solute carrier organic anion transporter family member 1B1 Homo sapiens 70-77 24194565-2 2014 Impaired hepatic clearance due to low-activity polymorphisms in human OATP1B1 may increase systemic exposure to SN-38, the active and toxic metabolite of the anticancer prodrug irinotecan. Irinotecan 177-187 solute carrier organic anion transporter family member 1B1 Homo sapiens 70-77 24701697-2 2014 Several recent phase III trials reported median overall survival data exceeding 30 months, an achievement inconceivable only 5 years ago.The first major step forward in the medical management of mCRC was provided by the addition of irinotecan and oxaliplatin to fluorouracil-based therapy; this increased survival from about 12 months to about 20 months.The introduction of biologic agents such as vascular endothelial growth factor inhibitors and epidermal growth factor inhibitors further increased survival--to more than 2 years in prospective trials. Irinotecan 232-242 vascular endothelial growth factor A Homo sapiens 398-432 24380837-0 2014 Optimization of irinotecan chronotherapy with P-glycoprotein inhibition. Irinotecan 16-26 phosphoglycolate phosphatase Mus musculus 46-60 24380837-1 2014 The relevance of P-glycoprotein (P-gp) for irinotecan chronopharmacology was investigated in female B6D2F1 mice. Irinotecan 43-53 phosphoglycolate phosphatase Mus musculus 33-37 24380837-7 2014 Non-tumor bearing mice lost an average of 17% or 9% of their body weight according to irinotecan administration at ZT3 or ZT15 respectively (p<0.001). Irinotecan 86-96 zinc finger protein 62 Mus musculus 115-118 24380837-12 2014 In conclusion, P-gp was an important determinant of the circadian balance between toxicity and efficacy of irinotecan. Irinotecan 107-117 phosphoglycolate phosphatase Mus musculus 15-19 24497922-1 2014 OBJECTIVE: To explore genes of the killer-cell immunoglobulin-like receptor (KIR) and of the HLA ligand and their relationship with the outcome of metastatic colorectal cancer (mCRC) patients treated with first-line 5-fluorouracil, leucovorin, and irinotecan (FOLFIRI). Irinotecan 248-258 killer cell immunoglobulin like receptor, two Ig domains and short cytoplasmic tail 4 Homo sapiens 77-80 24195516-2 2014 The metabolism of CPT-11 is mainly controlled by carboxy-lesterase (CES), UDP-glucuronosyltransferase 1A (UGT1A), and beta-glucuronidase (GUSB). Irinotecan 18-24 carboxylesterase 2 Homo sapiens 49-66 24815493-0 2014 Clinical observations on associations between the UGT1A1 genotype and severe toxicity of irinotecan. Irinotecan 89-99 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 50-56 24815493-2 2014 UDP glucuronosyltransferase1A1 (UGT1A1) catalyzes the glucuronidation of the active metabolite SN-38 but the relationship between UGT1A1 and severe toxicity remains unclear. Irinotecan 95-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-30 24815493-2 2014 UDP glucuronosyltransferase1A1 (UGT1A1) catalyzes the glucuronidation of the active metabolite SN-38 but the relationship between UGT1A1 and severe toxicity remains unclear. Irinotecan 95-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 32-38 24815493-2 2014 UDP glucuronosyltransferase1A1 (UGT1A1) catalyzes the glucuronidation of the active metabolite SN-38 but the relationship between UGT1A1 and severe toxicity remains unclear. Irinotecan 95-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 130-136 24815493-10 2014 CONCLUSIONS: Our study support the conclusion that patients with a UGT1A1*28 allele (s) will suffer an increased risk of severe irinotecan-induced diarrhea, whether with mid-or low-dosage. Irinotecan 128-138 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 67-73 24815493-12 2014 Higher serum total bilirubin is an indication that patients UGT1A1 genotype is not wild-type, with significance for clinic usage of CPT-11. Irinotecan 132-138 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 60-66 24172061-4 2014 In SW480 cells, DNMT inhibitors, such as decitabine (DAC), azacytidine and zebularine (Zeb), showed synergic effects on the cytotoxicity induced by anticancer drugs except for SN-38 plus Zeb, while HDAC inhibitors, trichostatin A, suberoylanilide hydroxamic acid and valproic acid, showed antagonistic effects. Irinotecan 176-181 DNA methyltransferase 1 Homo sapiens 16-20 24172061-4 2014 In SW480 cells, DNMT inhibitors, such as decitabine (DAC), azacytidine and zebularine (Zeb), showed synergic effects on the cytotoxicity induced by anticancer drugs except for SN-38 plus Zeb, while HDAC inhibitors, trichostatin A, suberoylanilide hydroxamic acid and valproic acid, showed antagonistic effects. Irinotecan 176-181 arylacetamide deacetylase Homo sapiens 53-56 24172061-8 2014 However, synergic effects of DAC with 5-FU or CPT-11 (SN-38) were not observed in 4 CRC cell lines. Irinotecan 54-59 arylacetamide deacetylase Homo sapiens 29-32 22454224-15 2014 Combined treatment of olaparib with irinotecan might be effective in treatment of non-BRCA-related breast cancer. Irinotecan 36-46 BRCA1 DNA repair associated Homo sapiens 86-90 25967674-0 2014 Prevalence of the UGT1A1*6 (c.211G>A) Polymorphism and Prediction of Irinotecan Toxicity in Iranian Populations of Different Ethnicities. Irinotecan 72-82 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 18-24 25967674-1 2014 BACKGROUND: Pharmacogenetic studies on irinotecan treatment in patients with metastatic colorectal cancer have indicated that genetic polymorphisms in UGT1A1*6 can lead to decreased enzyme activity and accumulation of the toxic metabolite SN-38. Irinotecan 39-49 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 151-157 25967674-1 2014 BACKGROUND: Pharmacogenetic studies on irinotecan treatment in patients with metastatic colorectal cancer have indicated that genetic polymorphisms in UGT1A1*6 can lead to decreased enzyme activity and accumulation of the toxic metabolite SN-38. Irinotecan 239-244 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 151-157 25967674-9 2014 CONCLUSION: The identification of the UGT1A1*6 alleles is necessary among the different Iranian ethnic groups before irinotecan therapy, suggesting that genotyping would be helpful for clinicians to optimize chemotherapy or identify individuals at risk of adverse drug reactions before clinical trials. Irinotecan 117-127 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 38-44 25016708-1 2014 BACKGROUND: Uridine diphosphate glucuronosyl transferase 1A1 (UGT1A1) is a key conjugating enzyme of bilirubin and the anti-tumor medication irinotecan. Irinotecan 141-151 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-60 25016708-1 2014 BACKGROUND: Uridine diphosphate glucuronosyl transferase 1A1 (UGT1A1) is a key conjugating enzyme of bilirubin and the anti-tumor medication irinotecan. Irinotecan 141-151 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 62-68 25016708-2 2014 Comprehensive analysis of UGT1A1 gene polymorphisms may provide benefit by predicting pharmacokinetics and outcomes of treatment with irinotecan and certain antiviral medications. Irinotecan 134-144 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 26-32 24988246-3 2014 Polymorphisms of the CES1 gene have been reported to affect the metabolism of dabigatran etexilate, methylphenidate, oseltamivir, imidapril, and clopidogrel, whereas variants of the CES2 gene have been found to affect aspirin and irinotecan. Irinotecan 230-240 carboxylesterase 1 Homo sapiens 21-25 24988246-3 2014 Polymorphisms of the CES1 gene have been reported to affect the metabolism of dabigatran etexilate, methylphenidate, oseltamivir, imidapril, and clopidogrel, whereas variants of the CES2 gene have been found to affect aspirin and irinotecan. Irinotecan 230-240 carboxylesterase 2 Homo sapiens 182-186 24173769-4 2014 Inhibition of NF-kappaB activation by p65 small interfering RNA was shown to potentiate apoptosis induced by CPT-11. Irinotecan 109-115 RELA proto-oncogene, NF-kB subunit Homo sapiens 38-41 24173769-5 2014 Berberine suppressed CPT-11-induced NF-kappaB activation in a dose-dependent manner and enhanced chemosensitivity to CPT-11 by downregulating NF-kappaB activation of antiapoptotic genes, c-IAP1, c-IAP2, survivin and Bcl-xL. Irinotecan 21-27 baculoviral IAP repeat containing 3 Homo sapiens 195-201 24173769-5 2014 Berberine suppressed CPT-11-induced NF-kappaB activation in a dose-dependent manner and enhanced chemosensitivity to CPT-11 by downregulating NF-kappaB activation of antiapoptotic genes, c-IAP1, c-IAP2, survivin and Bcl-xL. Irinotecan 21-27 BCL2 like 1 Homo sapiens 216-222 24195503-11 2014 Analysis of UGT1A1 alleles may be the basis for modified dosing and reducing the potential toxicity of irinotecan. Irinotecan 103-113 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-18 24368879-2 2014 Ziv-aflibercept is a fusion protein which acts as a decoy receptor for vascular endothelial growth factor (VEGF)-A, VEGF-B, and placental growth factor (PlGF); it was approved in combination with 5-fluorouracil, leucovorin, and irinotecan (FOLFIRI) for the treatment of patients with metastatic colorectal cancer that is resistant to or has progressed after an oxaliplatin-containing fluoropyrimidine-based regimen. Irinotecan 228-238 placental growth factor Homo sapiens 153-157 24594772-4 2014 The patient was treated with a combination of carboplatin and irinotecan, and achieved a partial response : size reduction of the prostate and the metastatic lesions, and decreased neuron specific enolase (NSE) level. Irinotecan 62-72 enolase 2 Homo sapiens 181-204 24594772-4 2014 The patient was treated with a combination of carboplatin and irinotecan, and achieved a partial response : size reduction of the prostate and the metastatic lesions, and decreased neuron specific enolase (NSE) level. Irinotecan 62-72 enolase 2 Homo sapiens 206-209 24190390-3 2014 In this study, we first demonstrate that a novel bFGF antagonist, peptide P7, previously isolated by phage display technology, reversed bFGF-induced resistance to irinotecan hydrochloride (CPT-11), and counteracted the anti-apoptotic effects of bFGF on CPT-11-treated HT-29 cells. Irinotecan 163-187 fibroblast growth factor 2 Homo sapiens 49-53 24190390-3 2014 In this study, we first demonstrate that a novel bFGF antagonist, peptide P7, previously isolated by phage display technology, reversed bFGF-induced resistance to irinotecan hydrochloride (CPT-11), and counteracted the anti-apoptotic effects of bFGF on CPT-11-treated HT-29 cells. Irinotecan 163-187 fibroblast growth factor 2 Homo sapiens 136-140 24190390-3 2014 In this study, we first demonstrate that a novel bFGF antagonist, peptide P7, previously isolated by phage display technology, reversed bFGF-induced resistance to irinotecan hydrochloride (CPT-11), and counteracted the anti-apoptotic effects of bFGF on CPT-11-treated HT-29 cells. Irinotecan 163-187 fibroblast growth factor 2 Homo sapiens 136-140 24190390-3 2014 In this study, we first demonstrate that a novel bFGF antagonist, peptide P7, previously isolated by phage display technology, reversed bFGF-induced resistance to irinotecan hydrochloride (CPT-11), and counteracted the anti-apoptotic effects of bFGF on CPT-11-treated HT-29 cells. Irinotecan 189-195 fibroblast growth factor 2 Homo sapiens 49-53 24190390-3 2014 In this study, we first demonstrate that a novel bFGF antagonist, peptide P7, previously isolated by phage display technology, reversed bFGF-induced resistance to irinotecan hydrochloride (CPT-11), and counteracted the anti-apoptotic effects of bFGF on CPT-11-treated HT-29 cells. Irinotecan 189-195 fibroblast growth factor 2 Homo sapiens 136-140 24190390-3 2014 In this study, we first demonstrate that a novel bFGF antagonist, peptide P7, previously isolated by phage display technology, reversed bFGF-induced resistance to irinotecan hydrochloride (CPT-11), and counteracted the anti-apoptotic effects of bFGF on CPT-11-treated HT-29 cells. Irinotecan 189-195 fibroblast growth factor 2 Homo sapiens 136-140 24190390-3 2014 In this study, we first demonstrate that a novel bFGF antagonist, peptide P7, previously isolated by phage display technology, reversed bFGF-induced resistance to irinotecan hydrochloride (CPT-11), and counteracted the anti-apoptotic effects of bFGF on CPT-11-treated HT-29 cells. Irinotecan 253-259 fibroblast growth factor 2 Homo sapiens 49-53 24190390-3 2014 In this study, we first demonstrate that a novel bFGF antagonist, peptide P7, previously isolated by phage display technology, reversed bFGF-induced resistance to irinotecan hydrochloride (CPT-11), and counteracted the anti-apoptotic effects of bFGF on CPT-11-treated HT-29 cells. Irinotecan 253-259 fibroblast growth factor 2 Homo sapiens 136-140 24190390-3 2014 In this study, we first demonstrate that a novel bFGF antagonist, peptide P7, previously isolated by phage display technology, reversed bFGF-induced resistance to irinotecan hydrochloride (CPT-11), and counteracted the anti-apoptotic effects of bFGF on CPT-11-treated HT-29 cells. Irinotecan 253-259 fibroblast growth factor 2 Homo sapiens 136-140 24190390-4 2014 Further experiments indicated that the inhibition of Akt activation, the suppression of bFGF internalization, the increase in the Bax to Bcl-2 ratio and the downregulation of cytokeratin 8 (CK8) by P7 may contribute to the counteracting of the anti-apoptotic effects of bFGF, and further reversal of bFGF-induced resistance to CPT-11. Irinotecan 327-333 keratin 8 Homo sapiens 190-193 24195516-2 2014 The metabolism of CPT-11 is mainly controlled by carboxy-lesterase (CES), UDP-glucuronosyltransferase 1A (UGT1A), and beta-glucuronidase (GUSB). Irinotecan 18-24 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 74-104 24195516-2 2014 The metabolism of CPT-11 is mainly controlled by carboxy-lesterase (CES), UDP-glucuronosyltransferase 1A (UGT1A), and beta-glucuronidase (GUSB). Irinotecan 18-24 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 106-111 24195516-2 2014 The metabolism of CPT-11 is mainly controlled by carboxy-lesterase (CES), UDP-glucuronosyltransferase 1A (UGT1A), and beta-glucuronidase (GUSB). Irinotecan 18-24 glucuronidase beta Homo sapiens 118-136 24195516-2 2014 The metabolism of CPT-11 is mainly controlled by carboxy-lesterase (CES), UDP-glucuronosyltransferase 1A (UGT1A), and beta-glucuronidase (GUSB). Irinotecan 18-24 glucuronidase beta Homo sapiens 138-142 24195516-11 2014 The promoter of the UGT1A1 gene in colorectal cancer cells is methylated, which is an important mechanism of UGT1A1 gene silencing and can be regarded as the target point of research for CPT-11 drug resistance and control mechanisms for the reversal of drug resistance. Irinotecan 187-193 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 20-26 24548447-15 2013 Patients with down-regulated ERCC1 on Oxaliplatin or up-regulated TOPO Ion Irinotecan have longer survival and better curative effect. Irinotecan 75-85 ERCC excision repair 1, endonuclease non-catalytic subunit Homo sapiens 29-34 24968756-3 2014 This prospective study investigated the relationship between the mutation status of EGFR-related genes including KRAS and the response rate (RR) to cetuximab plus irinotecan therapy in Japanese mCRC patients. Irinotecan 163-173 epidermal growth factor receptor Homo sapiens 84-88 24968756-3 2014 This prospective study investigated the relationship between the mutation status of EGFR-related genes including KRAS and the response rate (RR) to cetuximab plus irinotecan therapy in Japanese mCRC patients. Irinotecan 163-173 KRAS proto-oncogene, GTPase Homo sapiens 113-117 24968756-7 2014 The RR to cetuximab plus irinotecan therapy was found to be 17.9 and 0% in the KRAS wild-type and mutant subgroups, respectively. Irinotecan 25-35 KRAS proto-oncogene, GTPase Homo sapiens 79-83 24968756-8 2014 CONCLUSION: Despite the identification of a lower-than-expected RR to treatment by the KRAS wild-type subgroup, KRAS mutation status appears to be a useful predictive marker of response to cetuximab plus irinotecan therapy in Japanese mCRC patients. Irinotecan 204-214 KRAS proto-oncogene, GTPase Homo sapiens 112-116 23921491-7 2014 Kinetic analyses for OATP-mediated transport revealed that the uptake of SN-38 by OATP1B1 was the highest, followed by OATP1B3. Irinotecan 73-78 solute carrier organic anion transporter family member 1A2 Homo sapiens 21-25 23921491-7 2014 Kinetic analyses for OATP-mediated transport revealed that the uptake of SN-38 by OATP1B1 was the highest, followed by OATP1B3. Irinotecan 73-78 solute carrier organic anion transporter family member 1B1 Homo sapiens 82-89 23921491-7 2014 Kinetic analyses for OATP-mediated transport revealed that the uptake of SN-38 by OATP1B1 was the highest, followed by OATP1B3. Irinotecan 73-78 solute carrier organic anion transporter family member 1B3 Homo sapiens 119-126 23921491-8 2014 Among the uremic toxins, CMPF exhibited most potent inhibition of OATP1B1-mediated SN-38 uptake and directly inhibited the uptake of SN-38 also in hepatocytes. Irinotecan 83-88 solute carrier organic anion transporter family member 1B1 Homo sapiens 66-73 23921491-10 2014 CONCLUSIONS: OATP1B1-mediated hepatic uptake of SN-38 was inhibited by uremic toxins, and gene expression of OATP1B1 was down-regulated by uremic plasma. Irinotecan 48-53 solute carrier organic anion transporter family member 1B1 Homo sapiens 13-20 24359226-0 2013 Correlation between the methylation of SULF2 and WRN promoter and the irinotecan chemosensitivity in gastric cancer. Irinotecan 70-80 sulfatase 2 Homo sapiens 39-44 24359226-0 2013 Correlation between the methylation of SULF2 and WRN promoter and the irinotecan chemosensitivity in gastric cancer. Irinotecan 70-80 WRN RecQ like helicase Homo sapiens 49-52 24359226-7 2013 Patients with SULF2 methylation were more sensitive to CPT-11 than those without SULF2 methylation (P < 0.01). Irinotecan 55-61 sulfatase 2 Homo sapiens 14-19 24359226-8 2013 Patients with both SULF2 and WRN methylation were also more sensitive to CPT-11 than others (P < 0.05). Irinotecan 73-79 sulfatase 2 Homo sapiens 19-24 24359226-8 2013 Patients with both SULF2 and WRN methylation were also more sensitive to CPT-11 than others (P < 0.05). Irinotecan 73-79 WRN RecQ like helicase Homo sapiens 29-32 24359226-9 2013 CONCLUSION: SULF2 and WRN promoter methylation detection indicates potential predictive biomarkers to identify and target the most sensitive gastric cancer subpopulation for personalized CPT-11 therapy. Irinotecan 187-193 WRN RecQ like helicase Homo sapiens 22-25 24427321-11 2014 Curcumin may suppress GSTM5 expression to enhance the lethal effect of irinotecan on LOVO cells, and maybe their combination via the affection of PDI and PRDX4 to disturb the formation and reduction of disulfides results in inducing apoptosis of LOVO cell. Irinotecan 71-81 glutathione S-transferase mu 5 Homo sapiens 22-27 24154875-5 2013 The application of a maximum-a-posteriori Bayesian inference method identified a linear model based on Rev-erbalpha and Bmal1 circadian expressions that accurately predicted for optimal irinotecan timing. Irinotecan 186-196 nuclear receptor subfamily 1, group D, member 1 Mus musculus 103-115 24154875-5 2013 The application of a maximum-a-posteriori Bayesian inference method identified a linear model based on Rev-erbalpha and Bmal1 circadian expressions that accurately predicted for optimal irinotecan timing. Irinotecan 186-196 aryl hydrocarbon receptor nuclear translocator-like Mus musculus 120-125 24033692-1 2013 UNLABELLED: Retrospective studies have suggested that UDP-glucuronosyltransferase (UGT)1A1, UGT1A7, and UGT1A9 predict severe toxicity and efficacy of irinotecan-containing regimens. Irinotecan 151-161 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 83-90 24263065-0 2013 KRAS-mutated plasma DNA as predictor of outcome from irinotecan monotherapy in metastatic colorectal cancer. Irinotecan 53-63 KRAS proto-oncogene, GTPase Homo sapiens 0-4 24263065-1 2013 BACKGROUND: We investigated the clinical implications of KRAS and BRAF mutations detected in both archival tumor tissue and plasma cell-free DNA in metastatic colorectal cancer patients treated with irinotecan monotherapy. Irinotecan 199-209 KRAS proto-oncogene, GTPase Homo sapiens 57-61 24324067-3 2013 To apply this strategy to the treatment of CNS lymphoma, we investigated the role of NSCs expressing carboxyl esterase (HB1.F3.CE), which activates irinotecan. Irinotecan 148-158 histocompatibility minor HB-1 Homo sapiens 120-123 24324067-7 2013 RESULTS: The HB1.F3.CE cells significantly inhibited the growth of Raji cells with irinotecan treatment. Irinotecan 83-93 histocompatibility minor HB-1 Homo sapiens 13-16 24033692-1 2013 UNLABELLED: Retrospective studies have suggested that UDP-glucuronosyltransferase (UGT)1A1, UGT1A7, and UGT1A9 predict severe toxicity and efficacy of irinotecan-containing regimens. Irinotecan 151-161 UDP glucuronosyltransferase family 1 member A7 Homo sapiens 92-98 24033692-1 2013 UNLABELLED: Retrospective studies have suggested that UDP-glucuronosyltransferase (UGT)1A1, UGT1A7, and UGT1A9 predict severe toxicity and efficacy of irinotecan-containing regimens. Irinotecan 151-161 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 104-110 24311948-1 2013 BACKGROUND: The survival benefit for single-agent anti-epidermal growth factor receptor (egfr) therapy compared with combination therapy with irinotecan in KRAS wildtype (wt) metastatic colorectal cancer (mcrc) patients in the third-line treatment setting is not known. Irinotecan 142-152 KRAS proto-oncogene, GTPase Homo sapiens 156-160 24088669-0 2013 Polymorphisms in the UGT1A1 gene predict adverse effects of irinotecan in the treatment of gynecologic cancer in Japanese patients. Irinotecan 60-70 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 21-27 24379067-0 2013 PEG-coated irinotecan cationic liposomes improve the therapeutic efficacy of breast cancer in animals. Irinotecan 11-21 progestagen associated endometrial protein Homo sapiens 0-3 24379067-2 2013 AIM: This study aimed to construct a kind of PEG-coated irinotecan cationic liposomes for investigating its efficacy and mechanism of action in the treatment of breast cancer in preclinical models. Irinotecan 56-66 progestagen associated endometrial protein Homo sapiens 45-48 24379067-5 2013 RESULTS: The zeta potential of PEG-coated irinotecan cationic liposomes was approximately 23 mV. Irinotecan 42-52 progestagen associated endometrial protein Homo sapiens 31-34 24379067-6 2013 The PEG-coated irinotecan cationic liposomes were approximately 66nm in diameter, significantly increased the intracellular uptake of irinotecan, and showed strong inhibitory effect on MDA-MB231 breast cancer cells. Irinotecan 15-25 progestagen associated endometrial protein Homo sapiens 4-7 24379067-6 2013 The PEG-coated irinotecan cationic liposomes were approximately 66nm in diameter, significantly increased the intracellular uptake of irinotecan, and showed strong inhibitory effect on MDA-MB231 breast cancer cells. Irinotecan 134-144 progestagen associated endometrial protein Homo sapiens 4-7 24379067-7 2013 A significant antitumor efficacy in the xenografted MDA-MB231 breast cancer cells in nude mice was evidenced by intravenous administration of PEG-coated irinotecan cationic liposomes. Irinotecan 153-163 progestagen associated endometrial protein Homo sapiens 142-145 24379067-8 2013 PEG-coated irinotecan cationic liposomes also improved the irinotecan blood circulation time and showed an enhanced drug concentration in tumor. Irinotecan 11-21 progestagen associated endometrial protein Homo sapiens 0-3 24379067-8 2013 PEG-coated irinotecan cationic liposomes also improved the irinotecan blood circulation time and showed an enhanced drug concentration in tumor. Irinotecan 59-69 progestagen associated endometrial protein Homo sapiens 0-3 24379067-9 2013 CONCLUSIONS: PEG-coated irinotecan cationic liposomes had significant inhibitory effect against breast cancer in vitro and in vivo, hence providing a new strategy for treating breast cancer. Irinotecan 24-34 progestagen associated endometrial protein Homo sapiens 13-16 24114122-0 2013 UGT1A1 genotype-guided phase I study of irinotecan, oxaliplatin, and capecitabine. Irinotecan 40-50 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 24114122-11 2013 UGT1A1 *1/*28 and *1/*1 patients treated with IRIN (150) had similar AUCs for the active irinotecan metabolite, SN38 (366 +/- 278 and 350 +/- 159 ng/ml*hr, respectively). Irinotecan 89-99 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 24088669-6 2013 Homozygosity for UGT1A1*6/*6 and heterozygosity for UGT1A1*6/*28 were associated with an increased risk of absolute neutrophil count and/or diarrhea in Japanese gynecologic cancer patients, despite the lower doses of irinotecan used in these patients. Irinotecan 217-227 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 52-58 24088669-7 2013 UGT1A1*6 and UGT1A1*28 are potential predictors of severe absolute neutrophil decrease and diarrhea caused by low-dose irinotecan in gynecologic cancer patients. Irinotecan 119-129 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 24088669-7 2013 UGT1A1*6 and UGT1A1*28 are potential predictors of severe absolute neutrophil decrease and diarrhea caused by low-dose irinotecan in gynecologic cancer patients. Irinotecan 119-129 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 13-19 24088669-1 2013 Irinotecan is a key chemotherapeutic drug used to treat many tumors, including cervical and ovarian cancers; however, irinotecan can cause toxicity, particularly in the presence of uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) gene polymorphisms, which are associated with reduced enzyme activity. Irinotecan 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 230-236 24088669-1 2013 Irinotecan is a key chemotherapeutic drug used to treat many tumors, including cervical and ovarian cancers; however, irinotecan can cause toxicity, particularly in the presence of uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) gene polymorphisms, which are associated with reduced enzyme activity. Irinotecan 118-128 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 230-236 24088669-2 2013 Here, we investigated the prevalence of three different variants of UGT1A1 (UGT1A1*6, UGT1A1*27 and UGT1A1*28) and their relationships with irinotecan-induced adverse events in patients with gynecologic cancer, who are treated with lower doses of irinotecan than patients with other types of solid tumors. Irinotecan 140-150 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 68-74 24088669-2 2013 Here, we investigated the prevalence of three different variants of UGT1A1 (UGT1A1*6, UGT1A1*27 and UGT1A1*28) and their relationships with irinotecan-induced adverse events in patients with gynecologic cancer, who are treated with lower doses of irinotecan than patients with other types of solid tumors. Irinotecan 247-257 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 68-74 23394826-14 2013 In clinical studies using midazolam or anticancer drugs (irinotecan and imatinib) as known CYP3A4 substrates in combination with SJW, decreased plasma levels of these drugs were observed, which was expected as a consequence of CYP3A4 induction. Irinotecan 57-67 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 91-97 24132143-11 2013 We conclude that targeting ACLy may improve the therapeutic effects of irinotecan and that simultaneous targeting of ACLy and AKT may be warranted to overcome SN38 resistance. Irinotecan 71-81 ATP citrate lyase Homo sapiens 27-31 24129235-0 2013 A c-Met inhibitor increases the chemosensitivity of cancer stem cells to the irinotecan in gastric carcinoma. Irinotecan 77-87 MET proto-oncogene, receptor tyrosine kinase Homo sapiens 2-7 24129235-11 2013 The c-Met inhibitor may be a promising target molecule for irinotecan-based chemotherapy of gastric cancer. Irinotecan 59-69 MET proto-oncogene, receptor tyrosine kinase Homo sapiens 4-9 24081946-1 2013 PURPOSE: Etirinotecan pegol (NKTR-102) is a unique, long-acting topoisomerase-I inhibitor with prolonged systemic exposure to SN38 (7-ethyl-10-hydroxycamptothecin), the active metabolite of irinotecan. Irinotecan 126-130 natural killer cell triggering receptor Homo sapiens 29-33 24081946-1 2013 PURPOSE: Etirinotecan pegol (NKTR-102) is a unique, long-acting topoisomerase-I inhibitor with prolonged systemic exposure to SN38 (7-ethyl-10-hydroxycamptothecin), the active metabolite of irinotecan. Irinotecan 132-162 natural killer cell triggering receptor Homo sapiens 29-33 24081946-1 2013 PURPOSE: Etirinotecan pegol (NKTR-102) is a unique, long-acting topoisomerase-I inhibitor with prolonged systemic exposure to SN38 (7-ethyl-10-hydroxycamptothecin), the active metabolite of irinotecan. Irinotecan 11-21 natural killer cell triggering receptor Homo sapiens 29-33 24191041-3 2013 CPT-11 is a prodrug that is hydrolyzed by hepatic and intestinal carboxylesterase to form SN-38, which in turn is detoxified primarily through UDP-glucuronosyltransferase 1A1 (UGT1A1)-catalyzed glucuronidation. Irinotecan 0-6 UDP glucuronosyltransferase 1 family, polypeptide A1 Mus musculus 143-174 24191041-3 2013 CPT-11 is a prodrug that is hydrolyzed by hepatic and intestinal carboxylesterase to form SN-38, which in turn is detoxified primarily through UDP-glucuronosyltransferase 1A1 (UGT1A1)-catalyzed glucuronidation. Irinotecan 0-6 UDP glucuronosyltransferase 1 family, polypeptide A1 Mus musculus 176-182 24191041-3 2013 CPT-11 is a prodrug that is hydrolyzed by hepatic and intestinal carboxylesterase to form SN-38, which in turn is detoxified primarily through UDP-glucuronosyltransferase 1A1 (UGT1A1)-catalyzed glucuronidation. Irinotecan 90-95 UDP glucuronosyltransferase 1 family, polypeptide A1 Mus musculus 143-174 24191041-3 2013 CPT-11 is a prodrug that is hydrolyzed by hepatic and intestinal carboxylesterase to form SN-38, which in turn is detoxified primarily through UDP-glucuronosyltransferase 1A1 (UGT1A1)-catalyzed glucuronidation. Irinotecan 90-95 UDP glucuronosyltransferase 1 family, polypeptide A1 Mus musculus 176-182 24191041-8 2013 CPT-11-treated Ugt1(DeltaHep) mice showed a similar lethality rate to the CPT-11-treated Ugt1(F/F) mice. Irinotecan 0-6 UDP glucuronosyltransferase 1 family, polypeptide A2 Mus musculus 15-19 24191041-8 2013 CPT-11-treated Ugt1(DeltaHep) mice showed a similar lethality rate to the CPT-11-treated Ugt1(F/F) mice. Irinotecan 74-80 UDP glucuronosyltransferase 1 family, polypeptide A2 Mus musculus 89-93 24191041-9 2013 However, Ugt1(DeltaGI) mice were highly susceptible to CPT-11-induced diarrhea, developing severe and lethal mucositis at much lower CPT-11 doses, a result of the proliferative cell loss and inflammation in the intestinal tract. Irinotecan 55-61 UDP glucuronosyltransferase 1 family, polypeptide A2 Mus musculus 9-13 24191041-9 2013 However, Ugt1(DeltaGI) mice were highly susceptible to CPT-11-induced diarrhea, developing severe and lethal mucositis at much lower CPT-11 doses, a result of the proliferative cell loss and inflammation in the intestinal tract. Irinotecan 133-139 UDP glucuronosyltransferase 1 family, polypeptide A2 Mus musculus 9-13 24191041-11 2013 Intestinal expression of the UGT1A proteins is critical toward the detoxification of SN-38, whereas induction of the UGT1A1 gene may serve to limit toxicity and improve the efficacy associated with CPT-11 treatment. Irinotecan 85-90 Ugt1a@ Mus musculus 29-34 24191041-11 2013 Intestinal expression of the UGT1A proteins is critical toward the detoxification of SN-38, whereas induction of the UGT1A1 gene may serve to limit toxicity and improve the efficacy associated with CPT-11 treatment. Irinotecan 198-204 UDP glucuronosyltransferase 1 family, polypeptide A1 Mus musculus 117-123 23394826-14 2013 In clinical studies using midazolam or anticancer drugs (irinotecan and imatinib) as known CYP3A4 substrates in combination with SJW, decreased plasma levels of these drugs were observed, which was expected as a consequence of CYP3A4 induction. Irinotecan 57-67 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 227-233 23850745-1 2013 Irinotecan is a camptothecin derivative with low oral bioavailability due to active efflux by intestinal P-glycoprotein receptors. Irinotecan 0-10 ATP binding cassette subfamily B member 1 Homo sapiens 105-119 23850745-3 2013 However, an optimized Self micro emulsifying drug delivery system (SMEDDS), formulated to produce nano range oil droplets by using P-gp modulator excipients can tackle the issue and elevate the systemic availability of Irinotecan. Irinotecan 219-229 phosphoglycolate phosphatase Homo sapiens 131-135 23953030-3 2013 The canalicular transport of irinotecan and SN-38G is mediated by ABCC2 (MRP2) and ABCB1 (MDR1) which are both inhibited by ciclosporin. Irinotecan 29-39 ATP binding cassette subfamily C member 2 Homo sapiens 66-71 23953030-3 2013 The canalicular transport of irinotecan and SN-38G is mediated by ABCC2 (MRP2) and ABCB1 (MDR1) which are both inhibited by ciclosporin. Irinotecan 29-39 ATP binding cassette subfamily C member 2 Homo sapiens 73-77 23953030-3 2013 The canalicular transport of irinotecan and SN-38G is mediated by ABCC2 (MRP2) and ABCB1 (MDR1) which are both inhibited by ciclosporin. Irinotecan 29-39 ATP binding cassette subfamily B member 1 Homo sapiens 83-88 23953030-3 2013 The canalicular transport of irinotecan and SN-38G is mediated by ABCC2 (MRP2) and ABCB1 (MDR1) which are both inhibited by ciclosporin. Irinotecan 29-39 ATP binding cassette subfamily B member 1 Homo sapiens 90-94 23953030-3 2013 The canalicular transport of irinotecan and SN-38G is mediated by ABCC2 (MRP2) and ABCB1 (MDR1) which are both inhibited by ciclosporin. Irinotecan 44-49 ATP binding cassette subfamily C member 2 Homo sapiens 66-71 23953030-3 2013 The canalicular transport of irinotecan and SN-38G is mediated by ABCC2 (MRP2) and ABCB1 (MDR1) which are both inhibited by ciclosporin. Irinotecan 44-49 ATP binding cassette subfamily C member 2 Homo sapiens 73-77 23953030-3 2013 The canalicular transport of irinotecan and SN-38G is mediated by ABCC2 (MRP2) and ABCB1 (MDR1) which are both inhibited by ciclosporin. Irinotecan 44-49 ATP binding cassette subfamily B member 1 Homo sapiens 83-88 24231711-2 2013 The patient was treated with a pancreaticoduodenectomy, ascending colectomy and postoperative chemotherapy of S-1 and irinotecan[IRIS; S-1, 80mg/m2(days 1-14); irinotecan, 100 mg/m2(days 1 and 15, every 4 weeks)]. Irinotecan 118-128 proteasome 26S subunit, non-ATPase 1 Homo sapiens 135-138 23953030-3 2013 The canalicular transport of irinotecan and SN-38G is mediated by ABCC2 (MRP2) and ABCB1 (MDR1) which are both inhibited by ciclosporin. Irinotecan 44-49 ATP binding cassette subfamily B member 1 Homo sapiens 90-94 24124496-10 2013 SULF2 methylation was found to have a significant association with cisplatin efficacy(SULF2M: 57.14%, SULF2U: 80.56%, P = 0.02) and irinotecan efficacy(SULF2M: 89.29%, SULF2U: 62.50%, P = 0.01). Irinotecan 132-142 sulfatase 2 Homo sapiens 0-5 24038068-7 2013 We finally compared the ability of a structurally related CHK1 inhibitor, GNE-900, to enhance the in vivo activity of gemcitabine, CPT-11, and temozolomide in xenograft models. Irinotecan 131-137 checkpoint kinase 1 Homo sapiens 58-62 24038068-7 2013 We finally compared the ability of a structurally related CHK1 inhibitor, GNE-900, to enhance the in vivo activity of gemcitabine, CPT-11, and temozolomide in xenograft models. Irinotecan 131-137 glucosamine (UDP-N-acetyl)-2-epimerase/N-acetylmannosamine kinase Homo sapiens 74-77 23960095-0 2013 Sorafenib overcomes irinotecan resistance in colorectal cancer by inhibiting the ABCG2 drug-efflux pump. Irinotecan 20-30 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 81-86 23401472-1 2013 Human uridine 5"-diphospho-glucuronosyltransferase (UGT) 1A1 catalyzes the metabolism of numerous clinically and pharmacologically important compounds, such as bilirubin and SN-38. Irinotecan 174-179 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 6-60 22882086-2 2013 ABCG2 (BCRP) is an efflux transporter conferring cross-resistance to mitoxantrone (Mit), irinotecan (CPT11), and its active metabolite SN38. Irinotecan 89-99 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 0-5 22882086-2 2013 ABCG2 (BCRP) is an efflux transporter conferring cross-resistance to mitoxantrone (Mit), irinotecan (CPT11), and its active metabolite SN38. Irinotecan 89-99 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 7-11 22882086-2 2013 ABCG2 (BCRP) is an efflux transporter conferring cross-resistance to mitoxantrone (Mit), irinotecan (CPT11), and its active metabolite SN38. Irinotecan 101-106 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 0-5 22882086-2 2013 ABCG2 (BCRP) is an efflux transporter conferring cross-resistance to mitoxantrone (Mit), irinotecan (CPT11), and its active metabolite SN38. Irinotecan 101-106 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 7-11 22882086-2 2013 ABCG2 (BCRP) is an efflux transporter conferring cross-resistance to mitoxantrone (Mit), irinotecan (CPT11), and its active metabolite SN38. Irinotecan 135-139 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 0-5 22882086-2 2013 ABCG2 (BCRP) is an efflux transporter conferring cross-resistance to mitoxantrone (Mit), irinotecan (CPT11), and its active metabolite SN38. Irinotecan 135-139 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 7-11 24196082-8 2013 In November 2011, we started third-line chemotherapy using S-1/irinotecan (IRIS) with PSK. Irinotecan 63-73 TAO kinase 2 Homo sapiens 86-89 23913164-5 2013 Critically, knockdown of TDP1 or inhibition of poly(ADP-ribose) polymerase-1 (PARP-1), an enzyme in the same complex as TDP1, sensitized rhabdomyosarcoma cell lines to analogues of irinotecan. Irinotecan 181-191 tyrosyl-DNA phosphodiesterase 1 Homo sapiens 25-29 23913164-5 2013 Critically, knockdown of TDP1 or inhibition of poly(ADP-ribose) polymerase-1 (PARP-1), an enzyme in the same complex as TDP1, sensitized rhabdomyosarcoma cell lines to analogues of irinotecan. Irinotecan 181-191 poly(ADP-ribose) polymerase 1 Homo sapiens 47-76 23913164-5 2013 Critically, knockdown of TDP1 or inhibition of poly(ADP-ribose) polymerase-1 (PARP-1), an enzyme in the same complex as TDP1, sensitized rhabdomyosarcoma cell lines to analogues of irinotecan. Irinotecan 181-191 poly(ADP-ribose) polymerase 1 Homo sapiens 78-84 23913164-5 2013 Critically, knockdown of TDP1 or inhibition of poly(ADP-ribose) polymerase-1 (PARP-1), an enzyme in the same complex as TDP1, sensitized rhabdomyosarcoma cell lines to analogues of irinotecan. Irinotecan 181-191 tyrosyl-DNA phosphodiesterase 1 Homo sapiens 120-124 23960095-2 2013 Therefore, new treatment options are needed to improve survival of patients with irinotecan-refractory CRCs, particularly those bearing KRAS mutations that preclude the use of anti-EGFR therapies. Irinotecan 81-91 KRAS proto-oncogene, GTPase Homo sapiens 136-140 23960095-2 2013 Therefore, new treatment options are needed to improve survival of patients with irinotecan-refractory CRCs, particularly those bearing KRAS mutations that preclude the use of anti-EGFR therapies. Irinotecan 81-91 epidermal growth factor receptor Homo sapiens 181-185 23960095-6 2013 By inhibiting the drug-efflux pump ABCG2, sorafenib favors irinotecan intracellular accumulation and enhances its toxicity. Irinotecan 59-69 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 35-40 23960095-7 2013 Moreover, we found that sorafenib improved the efficacy of irinotecan by inhibiting the irinotecan-mediated p38 and ERK activation. Irinotecan 59-69 mitogen-activated protein kinase 14 Homo sapiens 108-111 23960095-7 2013 Moreover, we found that sorafenib improved the efficacy of irinotecan by inhibiting the irinotecan-mediated p38 and ERK activation. Irinotecan 59-69 mitogen-activated protein kinase 1 Homo sapiens 116-119 23960095-7 2013 Moreover, we found that sorafenib improved the efficacy of irinotecan by inhibiting the irinotecan-mediated p38 and ERK activation. Irinotecan 88-98 mitogen-activated protein kinase 14 Homo sapiens 108-111 23960095-7 2013 Moreover, we found that sorafenib improved the efficacy of irinotecan by inhibiting the irinotecan-mediated p38 and ERK activation. Irinotecan 88-98 mitogen-activated protein kinase 1 Homo sapiens 116-119 24069296-0 2013 Prognostic value of CD109+ circulating endothelial cells in recurrent glioblastomas treated with bevacizumab and irinotecan. Irinotecan 113-123 CD109 molecule Homo sapiens 20-25 24018773-2 2013 The aim of this study was to elucidate the predictive/prognostic role of ATP-binding cassette (ABC) and solute carrier (SLC) protein polymorphisms on irinotecan (FOLFIRI regimen) outcome. Irinotecan 150-160 ATP binding cassette subfamily B member 6 (Langereis blood group) Homo sapiens 73-93 24018773-2 2013 The aim of this study was to elucidate the predictive/prognostic role of ATP-binding cassette (ABC) and solute carrier (SLC) protein polymorphisms on irinotecan (FOLFIRI regimen) outcome. Irinotecan 150-160 ATP binding cassette subfamily B member 6 (Langereis blood group) Homo sapiens 95-98 24069296-7 2013 Patients treated with bevacizumab with or without irinotecan that were free from MRI progression after two months of treatment had significant decrease of CD109+ CECs: median PFS was 19 weeks; median OS 29 weeks. Irinotecan 50-60 CD109 molecule Homo sapiens 155-160 23859194-6 2013 We report that the DNA-damaging agents doxorubicin and 7-ethyl-10-hydroxycamptothecin were able to induce the formation of SAHF in some tumour cell types, and this induction was accompanied by activation of the retinoblastoma protein pathway. Irinotecan 55-85 RB transcriptional corepressor 1 Homo sapiens 211-225 23835420-5 2013 OCT1-mediated morphine uptake was abolished by common loss-of-function polymorphisms in the OCT1 gene and was strongly inhibited by drug-drug interactions with irinotecan, verapamil and ondansetron. Irinotecan 160-170 solute carrier family 22 member 1 Homo sapiens 0-4 23835420-5 2013 OCT1-mediated morphine uptake was abolished by common loss-of-function polymorphisms in the OCT1 gene and was strongly inhibited by drug-drug interactions with irinotecan, verapamil and ondansetron. Irinotecan 160-170 solute carrier family 22 member 1 Homo sapiens 92-96 23359181-0 2013 Panitumumab and irinotecan every 3 weeks is an active and convenient regimen for second-line treatment of patients with wild-type K-RAS metastatic colorectal cancer. Irinotecan 16-26 KRAS proto-oncogene, GTPase Homo sapiens 130-135 23359181-1 2013 PURPOSE: To evaluate the efficacy and safety profile of the combination of panitumumab and irinotecan every 3 weeks in a phase II trial as second-line treatment in patients with advanced wild-type (WT) K-RAS colorectal cancer (CRC). Irinotecan 91-101 KRAS proto-oncogene, GTPase Homo sapiens 202-207 23359181-10 2013 CONCLUSION: This study shows that the administration of panitumumab plus irinotecan every 3 weeks is safe, active and feasible as second-line treatment in patients with advanced WT K-RAS CRC. Irinotecan 73-83 KRAS proto-oncogene, GTPase Homo sapiens 181-186 23978462-1 2013 BACKGROUND: Kirsten rat sarcoma virus (KRAS) wild-type status determined using a locked nucleic acid (LNA)-mediated quantitative polymerase chain reaction (qPCR) clamping assay (LNA assay) predicted response to therapy in the CRYSTAL (Cetuximab Combined With Irinotecan in First-Line Therapy for Metastatic Colorectal Cancer) study. Irinotecan 259-269 KRAS proto-oncogene, GTPase Homo sapiens 39-43 23859194-9 2013 In addition, we show that the mitogen-activated protein kinase p38 pathway was involved in tumour cell SAHF formation in response to doxorubicin and 7-ethyl-10-hydroxycamptothecin. Irinotecan 149-179 mitogen-activated protein kinase 14 Homo sapiens 63-66 22643568-2 2013 In this study, we evaluated if there is any difference between first-line irinotecan-based and oxaliplatin-based chemotherapies in the duration of time to disease progression (TTP) in CRC patients with only liver metastasis. Irinotecan 74-84 ZFP36 ring finger protein Homo sapiens 176-179 23892411-0 2013 Analysis of UGT1A1*28 genotype and SN-38 pharmacokinetics for irinotecan-based chemotherapy in patients with advanced colorectal cancer: results from a multicenter, retrospective study in Shanghai. Irinotecan 62-72 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-18 23892411-1 2013 BACKGROUND: The UGT1A1*28 polymorphism, although closely linked with CPT-11-related adverse effects, cannot be used alone to guide individualized treatment decisions. Irinotecan 69-75 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 16-22 22643568-7 2013 TTP was significantly improved for patients who received irinotecan+5-FU+bevacizumab (median TTP, 13.73 +- 2.10 mo) when compared with irinotecan+5-FU (median TTP, 5.13 +- 0.70 mo). Irinotecan 57-67 ZFP36 ring finger protein Homo sapiens 0-3 22643568-7 2013 TTP was significantly improved for patients who received irinotecan+5-FU+bevacizumab (median TTP, 13.73 +- 2.10 mo) when compared with irinotecan+5-FU (median TTP, 5.13 +- 0.70 mo). Irinotecan 57-67 ZFP36 ring finger protein Homo sapiens 93-96 22643568-7 2013 TTP was significantly improved for patients who received irinotecan+5-FU+bevacizumab (median TTP, 13.73 +- 2.10 mo) when compared with irinotecan+5-FU (median TTP, 5.13 +- 0.70 mo). Irinotecan 57-67 ZFP36 ring finger protein Homo sapiens 93-96 23898114-0 2013 UDP-glucuronosyltransferase (UGT) 1A1*28 polymorphism-directed phase II study of irinotecan with 5"-deoxy-5-fluorouridine (5"-DFUR) for metastatic colorectal cancer. Irinotecan 81-91 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-37 23898114-1 2013 AIM: We performed a phase II study of irinotecan with 5"-deoxy-5-fluorouridine (5"-DFUR) for metastatic colorectal cancer based on UDP-glucuronosyltransferase (UGT) 1A1 polymorphism. Irinotecan 38-48 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 131-168 23898097-4 2013 In contrast, patient cohorts treated with irinotecan had significantly worse mOS (10.20 vs. 13.55 months, p=0.008, 35 vs. 810 cohorts) and a disadvantage compared to cohorts treated without irinotecan in survival gain analysis. Irinotecan 42-52 Moloney sarcoma oncogene Mus musculus 77-80 23800356-0 2013 Recurrent cervical cancer in a patient who was compound heterozygous for UGT1A1*6 and UGT1A1*28 presenting with serious adverse events during irinotecan hydrochloride/nedaplatin therapy. Irinotecan 142-166 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 73-79 24156021-7 2013 Finally, we evaluated enhancement of cytotoxicity of S-1 by CPT-11 (7-ethyl-10-[4-(1-piperidino)-1-piperidino]carbonyloxycamptothecin) which down-regulated TS in in vivo study. Irinotecan 60-66 proteasome 26S subunit, non-ATPase 1 Homo sapiens 53-56 24156021-7 2013 Finally, we evaluated enhancement of cytotoxicity of S-1 by CPT-11 (7-ethyl-10-[4-(1-piperidino)-1-piperidino]carbonyloxycamptothecin) which down-regulated TS in in vivo study. Irinotecan 60-66 thymidylate synthetase Homo sapiens 156-158 24156021-7 2013 Finally, we evaluated enhancement of cytotoxicity of S-1 by CPT-11 (7-ethyl-10-[4-(1-piperidino)-1-piperidino]carbonyloxycamptothecin) which down-regulated TS in in vivo study. Irinotecan 68-133 proteasome 26S subunit, non-ATPase 1 Homo sapiens 53-56 24156021-7 2013 Finally, we evaluated enhancement of cytotoxicity of S-1 by CPT-11 (7-ethyl-10-[4-(1-piperidino)-1-piperidino]carbonyloxycamptothecin) which down-regulated TS in in vivo study. Irinotecan 68-133 thymidylate synthetase Homo sapiens 156-158 24156021-10 2013 Down-regulation of TS was observed after exposure to SN-38 (7-ethyl-10-hydroxycamptothecin) in a dose-dependent manner. Irinotecan 53-58 thymidylate synthetase Homo sapiens 19-21 24156021-10 2013 Down-regulation of TS was observed after exposure to SN-38 (7-ethyl-10-hydroxycamptothecin) in a dose-dependent manner. Irinotecan 60-90 thymidylate synthetase Homo sapiens 19-21 24156021-13 2013 Also, combined S-1 and CPT-11 dramatically inhibited tumor growth, compared to S-1 or CPT-11 alone in in vivo study. Irinotecan 86-92 proteasome 26S subunit, non-ATPase 1 Homo sapiens 15-18 24156021-14 2013 In conclusion, CPT-11 down-regulated TS level and enhanced the effect of S-1. Irinotecan 15-21 thymidylate synthetase Homo sapiens 37-39 24156021-14 2013 In conclusion, CPT-11 down-regulated TS level and enhanced the effect of S-1. Irinotecan 15-21 proteasome 26S subunit, non-ATPase 1 Homo sapiens 73-76 24156021-15 2013 Thus, the combination therapy with S-1 and CPT-11 might be a novel modality for bladder cancer, even with high TS level. Irinotecan 43-49 thymidylate synthetase Homo sapiens 111-113 23852648-3 2013 The aim of this trial is to evaluate the efficacy and safety of combination chemotherapy with trastuzumab and irinotecan in Japanese patients with advanced human epidermal growth factor receptor 2-positive chemo-refractory gastric cancer. Irinotecan 110-120 erb-b2 receptor tyrosine kinase 2 Homo sapiens 162-196 23756919-8 2013 There was a strong correlation between hepatic CES-2 expression and activation of irinotecan (r (2) = 0.96, p < 0.001). Irinotecan 82-92 carboxylesterase 2 Homo sapiens 47-52 23756919-10 2013 Hepatic CES-2 mediated activation of irinotecan clearly correlated with tumour replacement by fibrosis (r (2) = 0.54, p = 0.01). Irinotecan 37-47 carboxylesterase 2 Homo sapiens 8-13 23800356-0 2013 Recurrent cervical cancer in a patient who was compound heterozygous for UGT1A1*6 and UGT1A1*28 presenting with serious adverse events during irinotecan hydrochloride/nedaplatin therapy. Irinotecan 142-166 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 86-92 23800356-6 2013 The findings in this patient indicate that CPT-11 should be administered with great care, even at a dose of 60 mg/m2, in patients receiving combined therapy with CPT-11 and NDP who are compound heterozygous for UGT1A1*6 and UGT1A1*28. Irinotecan 43-49 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 211-217 23800356-6 2013 The findings in this patient indicate that CPT-11 should be administered with great care, even at a dose of 60 mg/m2, in patients receiving combined therapy with CPT-11 and NDP who are compound heterozygous for UGT1A1*6 and UGT1A1*28. Irinotecan 43-49 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 224-230 23800356-6 2013 The findings in this patient indicate that CPT-11 should be administered with great care, even at a dose of 60 mg/m2, in patients receiving combined therapy with CPT-11 and NDP who are compound heterozygous for UGT1A1*6 and UGT1A1*28. Irinotecan 162-168 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 211-217 23690068-0 2013 Molecular insights into microbial beta-glucuronidase inhibition to abrogate CPT-11 toxicity. Irinotecan 76-82 glucuronidase beta Homo sapiens 34-52 24511364-2 2013 In the present study curcumin inhibitory action on p-glycoprotein increased permeability of irinotecan, so in the colon cancer it would be beneficial if curcumin used as add on therapy. Irinotecan 92-102 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 51-65 23180349-0 2013 Lack of Bax expression is associated with irinotecan-based treatment activity in advanced colorectal cancer patients. Irinotecan 42-52 BCL2 associated X, apoptosis regulator Homo sapiens 8-11 23180349-2 2013 Bax proapoptotic protein may correlate to chemosensitivity and differential response to irinotecan or oxaliplatin-based combinations. Irinotecan 88-98 BCL2 associated X, apoptosis regulator Homo sapiens 0-3 23180349-3 2013 METHODS: Bax expression was assessed by immunohistochemistry in 49 advanced colorectal cancer patients enrolled at our institution from 2002 to 2004 within a multicenter, phase II, randomized trial of first-line UFT/leucovorin/irinotecan (TEGAFIRI) versus UFT/leucovorin/oxaliplatin (TEGAFOX). Irinotecan 227-237 BCL2 associated X, apoptosis regulator Homo sapiens 9-12 23180349-8 2013 CONCLUSION: Bax-negative colorectal cancer may identify a specific phenotype of patients with significantly higher chance to respond to doublet irinotecan-based chemotherapy. Irinotecan 144-154 BCL2 associated X, apoptosis regulator Homo sapiens 12-15 27121781-1 2013 This study examined the effects of panitumumab, a human monoclonal antibody against epidermal growth factor receptor (EGFR), on irinotecan pharmacokinetics. Irinotecan 128-138 epidermal growth factor receptor Homo sapiens 84-116 23840132-0 2013 Severe irinotecan-induced toxicity in a patient with UGT1A1 28 and UGT1A1 6 polymorphisms. Irinotecan 7-17 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 53-59 27121781-1 2013 This study examined the effects of panitumumab, a human monoclonal antibody against epidermal growth factor receptor (EGFR), on irinotecan pharmacokinetics. Irinotecan 128-138 epidermal growth factor receptor Homo sapiens 118-122 22791234-2 2013 Irinotecan demonstrates single-agent activity in head and neck cancer but activates NF-kappaB, promoting cell survival and resistance. Irinotecan 0-10 nuclear factor kappa B subunit 1 Homo sapiens 84-93 23653048-0 2013 Influence of MLH1 on colon cancer sensitivity to poly(ADP-ribose) polymerase inhibitor combined with irinotecan. Irinotecan 101-111 mutL homolog 1 Homo sapiens 13-17 23653048-5 2013 The results of in vitro and in vivo experiments indicated that MLH1, together with low levels of Top1, contributed to colon cancer resistance to irinotecan. Irinotecan 145-155 mutL homolog 1 Homo sapiens 63-67 23653048-6 2013 In the MLH1-proficient cells SN-38, the active metabolite of irinotecan, induced lower levels of DNA damage than in MLH1-deficient cells, as shown by the weaker induction of gamma-H2AX and p53 phosphorylation. Irinotecan 29-34 mutL homolog 1 Homo sapiens 7-11 23653048-6 2013 In the MLH1-proficient cells SN-38, the active metabolite of irinotecan, induced lower levels of DNA damage than in MLH1-deficient cells, as shown by the weaker induction of gamma-H2AX and p53 phosphorylation. Irinotecan 29-34 tumor protein p53 Homo sapiens 189-192 23653048-6 2013 In the MLH1-proficient cells SN-38, the active metabolite of irinotecan, induced lower levels of DNA damage than in MLH1-deficient cells, as shown by the weaker induction of gamma-H2AX and p53 phosphorylation. Irinotecan 61-71 mutL homolog 1 Homo sapiens 7-11 23653048-6 2013 In the MLH1-proficient cells SN-38, the active metabolite of irinotecan, induced lower levels of DNA damage than in MLH1-deficient cells, as shown by the weaker induction of gamma-H2AX and p53 phosphorylation. Irinotecan 61-71 tumor protein p53 Homo sapiens 189-192 23653048-9 2013 Remarkably, inhibition of PARP function by PARPi or by PARP-1 gene silencing always increased the antitumor activity of irinotecan, even in the presence of low PARP-1 expression. Irinotecan 120-130 poly(ADP-ribose) polymerase 1 Homo sapiens 26-30 23653048-9 2013 Remarkably, inhibition of PARP function by PARPi or by PARP-1 gene silencing always increased the antitumor activity of irinotecan, even in the presence of low PARP-1 expression. Irinotecan 120-130 poly(ADP-ribose) polymerase 1 Homo sapiens 55-61 23653048-9 2013 Remarkably, inhibition of PARP function by PARPi or by PARP-1 gene silencing always increased the antitumor activity of irinotecan, even in the presence of low PARP-1 expression. Irinotecan 120-130 poly(ADP-ribose) polymerase 1 Homo sapiens 160-166 23583082-10 2013 In the PET study in mice, the radioactivity levels in the brain, liver, and small intestine were slightly increased by inhibition of the Pgp/Bcrp function for more than 30 min after [(11)C]irinotecan injection. Irinotecan 189-199 phosphoglycolate phosphatase Mus musculus 137-140 23583082-10 2013 In the PET study in mice, the radioactivity levels in the brain, liver, and small intestine were slightly increased by inhibition of the Pgp/Bcrp function for more than 30 min after [(11)C]irinotecan injection. Irinotecan 189-199 ATP binding cassette subfamily G member 2 (Junior blood group) Mus musculus 141-145 23583082-11 2013 This result demonstrated that in vivo behavior of [(11)C] irinotecan and radioactive metabolites are influenced by the Pgp/Bcrp function. Irinotecan 58-68 phosphoglycolate phosphatase Mus musculus 119-122 23583082-11 2013 This result demonstrated that in vivo behavior of [(11)C] irinotecan and radioactive metabolites are influenced by the Pgp/Bcrp function. Irinotecan 58-68 ATP binding cassette subfamily G member 2 (Junior blood group) Mus musculus 123-127 23126525-6 2013 The combination of melatonin at concentrations of 50 muM with increasing doses of irinotecan (7.5, 15, 30, and 60 muM) resulted in an increase in the amount of DNA damage in A549 and HT29 cancer cells, but was not effective in inducing DNA damage in healthy human lymphocytes. Irinotecan 82-92 latexin Homo sapiens 114-117 23420304-11 2013 The subgroup of patients, who received irinotecan had a higher complication rate in the CTX group than in the BV + CTX group (55 % vs 41 %). Irinotecan 39-49 cytochrome P450 family 27 subfamily A member 1 Homo sapiens 88-91 23420304-11 2013 The subgroup of patients, who received irinotecan had a higher complication rate in the CTX group than in the BV + CTX group (55 % vs 41 %). Irinotecan 39-49 cytochrome P450 family 27 subfamily A member 1 Homo sapiens 115-118 23840132-0 2013 Severe irinotecan-induced toxicity in a patient with UGT1A1 28 and UGT1A1 6 polymorphisms. Irinotecan 7-17 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 67-73 23840132-1 2013 Many studies have demonstrated the impact of UGT1A1 on toxicity of irinotecan. Irinotecan 67-77 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 45-51 23840132-3 2013 Based on this, prescribers of irinotecan are advised that patients with UGT1A1 28 (TA 7/7) should start with a reduced dose of irinotecan, although a particular dose is not specified. Irinotecan 30-40 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 72-78 23840132-3 2013 Based on this, prescribers of irinotecan are advised that patients with UGT1A1 28 (TA 7/7) should start with a reduced dose of irinotecan, although a particular dose is not specified. Irinotecan 127-137 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 72-78 23840132-4 2013 Research in Asian countries has shown a lower incidence of UGT1A1 28 (TA 7/7), while UGT1A1 6 (A/A) is more often found and is associated with severe irinotecan-related neutropenia. Irinotecan 150-160 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 85-91 23840132-7 2013 Simultaneous heterozygous UGT1A1 28 and UGT1A1 6 polymorphisms may produce higher exposure to SN-38 and a higher risk of adverse effects related to irinotecan. Irinotecan 94-99 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 26-32 23840132-7 2013 Simultaneous heterozygous UGT1A1 28 and UGT1A1 6 polymorphisms may produce higher exposure to SN-38 and a higher risk of adverse effects related to irinotecan. Irinotecan 94-99 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 40-46 23840132-7 2013 Simultaneous heterozygous UGT1A1 28 and UGT1A1 6 polymorphisms may produce higher exposure to SN-38 and a higher risk of adverse effects related to irinotecan. Irinotecan 148-158 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 26-32 23840132-7 2013 Simultaneous heterozygous UGT1A1 28 and UGT1A1 6 polymorphisms may produce higher exposure to SN-38 and a higher risk of adverse effects related to irinotecan. Irinotecan 148-158 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 40-46 23840132-8 2013 Additional studies will be necessary to determine the optimal starting dose of irinotecan for patients with both UGT1A1 28 and UGT1A1 6 polymorphisms. Irinotecan 79-89 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 113-119 23840132-8 2013 Additional studies will be necessary to determine the optimal starting dose of irinotecan for patients with both UGT1A1 28 and UGT1A1 6 polymorphisms. Irinotecan 79-89 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 127-133 23762305-9 2013 There were also weak but significant correlations between APTX mRNA expression levels and CPT-11 sensitivity (rho = -0.376, P = 0.017) or EVO sensitivity (rho = -0.322, P = 0.036). Irinotecan 90-96 aprataxin Homo sapiens 58-62 23514584-0 2013 Phase II trial of temsirolimus alone and in combination with irinotecan for KRAS mutant metastatic colorectal cancer: outcome and results of KRAS mutational analysis in plasma. Irinotecan 61-71 KRAS proto-oncogene, GTPase Homo sapiens 76-80 23595344-0 2013 A UGT1A1*28 and *6 genotype-directed phase I dose-escalation trial of irinotecan with fixed-dose capecitabine in Korean patients with metastatic colorectal cancer. Irinotecan 70-80 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 2-8 22721392-0 2013 The role of autophagic cell death and apoptosis in irinotecan-treated p53 null colon cancer cells. Irinotecan 51-61 tumor protein p53 Homo sapiens 70-73 22721392-1 2013 The roles of autophagic cell death and apoptosis induced by topoisomerase inhibitor irinotecan in colon cancer cells with deleted p53 were investigated during 48 h. We report that irinotecan-dependent cytotoxicity and proapoptotic activity were reduced in the present model while autophagy levels significantly increased. Irinotecan 180-190 tumor protein p53 Homo sapiens 130-133 22721392-4 2013 These results suggest that different modes of cell death in p53 null colon cancer cells treated with cytostatics (irinotecan) may be activated simultaneously. Irinotecan 114-124 tumor protein p53 Homo sapiens 60-63 23595344-10 2013 CONCLUSION: Irinotecan dosing based on UGT1A1*28 and *6 is feasible, and higher doses of irinotecan can be safely administered in patients with 0 or 1 DA, compared to those with 2 DA. Irinotecan 12-22 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 39-45 23595344-1 2013 PURPOSE: UGT1A1 genotypes are important when considering treatment with irinotecan-containing regimens. Irinotecan 72-82 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 9-15 23595344-2 2013 In this study, we determined the dose, efficacy, and tolerability of irinotecan according to UGT1A1 genotypes when combined with capecitabine in patients with metastatic colorectal cancer. Irinotecan 69-79 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-99 23427296-9 2013 Despite much lower surface expression of CD74 than Trop-2 or CEACAM6, milatuzumab-SN-38 had similar efficacy in Capan-1 as anti-Trop-2-SN-38, but in NCI-N87, anti-CEACAM6 and anti-Trop-2 conjugates were superior. Irinotecan 82-87 CEA cell adhesion molecule 6 Homo sapiens 163-170 23427296-11 2013 CD74 is a suitable target for ADCs in some solid tumor xenografts, with efficacy largely influenced by uniformity of CD74 expression and with SN-38 conjugates providing the best therapeutic responses; SN-38 conjugates were preferable in solid cancers, whereas doxorubicin ADC was better in lymphoma tested. Irinotecan 142-147 CD74 molecule Homo sapiens 0-4 23427296-11 2013 CD74 is a suitable target for ADCs in some solid tumor xenografts, with efficacy largely influenced by uniformity of CD74 expression and with SN-38 conjugates providing the best therapeutic responses; SN-38 conjugates were preferable in solid cancers, whereas doxorubicin ADC was better in lymphoma tested. Irinotecan 201-206 CD74 molecule Homo sapiens 0-4 23801203-0 2013 [Correlation of MDR1 and ABCG2 genetic polymorphisms with the efficacy and adverse events of irinotecan chemotherapy in patients with colorectal cancer]. Irinotecan 93-103 ATP binding cassette subfamily B member 1 Homo sapiens 16-20 23859115-0 2013 Cetuximab/irinotecan-chemotherapy in KRAS wild-type pretreated metastatic colorectal cancer: a pooled analysis and review of literature. Irinotecan 10-20 KRAS proto-oncogene, GTPase Homo sapiens 37-41 23859115-11 2013 CONCLUSION: In metastatic KRAS wild-type CRC patients pretreated with one or more lines of therapy, cetuximab plus irinotecan-based chemotherapy is an active treatment. Irinotecan 115-125 KRAS proto-oncogene, GTPase Homo sapiens 26-30 23730417-8 2013 In contrast, in patients who received first-line irinotecan-based regimens, the PFS was shorter in KRAS mutant patients (N = 15) than that in KRAS wild-type patients (N = 20). Irinotecan 49-59 KRAS proto-oncogene, GTPase Homo sapiens 99-103 23730417-8 2013 In contrast, in patients who received first-line irinotecan-based regimens, the PFS was shorter in KRAS mutant patients (N = 15) than that in KRAS wild-type patients (N = 20). Irinotecan 49-59 KRAS proto-oncogene, GTPase Homo sapiens 142-146 24119900-0 2013 [Correlation between the methylation of SULF2 and WRN promoter and chemosensitivity to irinotecan in gastric cancer]. Irinotecan 87-97 sulfatase 2 Homo sapiens 40-45 24119900-0 2013 [Correlation between the methylation of SULF2 and WRN promoter and chemosensitivity to irinotecan in gastric cancer]. Irinotecan 87-97 WRN RecQ like helicase Homo sapiens 50-53 24119900-1 2013 OBJECTIVE: To explore the relationship between SULF2 and WRN promoter methylation and chemosensitivity to irinotecan, and also the clinicopathological features in patients with gastric cancer. Irinotecan 106-116 sulfatase 2 Homo sapiens 47-52 24119900-1 2013 OBJECTIVE: To explore the relationship between SULF2 and WRN promoter methylation and chemosensitivity to irinotecan, and also the clinicopathological features in patients with gastric cancer. Irinotecan 106-116 WRN RecQ like helicase Homo sapiens 57-60 24119900-11 2013 The nude mice bearing human gastric cancer xenografts with SULF2 methylation were more sensitive to irrinotecan. Irinotecan 100-111 sulfatase 2 Homo sapiens 59-64 24119900-12 2013 CONCLUSIONS: The detection of SULF2 and WRN promoter methylation may provide evidence for screening and targeting the most sensitive gastric cancer subpopulation suitable for personalized irrinotecan chemotherapy. Irinotecan 188-199 sulfatase 2 Homo sapiens 30-35 24119900-12 2013 CONCLUSIONS: The detection of SULF2 and WRN promoter methylation may provide evidence for screening and targeting the most sensitive gastric cancer subpopulation suitable for personalized irrinotecan chemotherapy. Irinotecan 188-199 WRN RecQ like helicase Homo sapiens 40-43 23801203-12 2013 CONCLUSION: MDR1 2677 G>T/A may be served as a biomarker in predicting the efficacy of irinotecan chemotherapy in patients with colorectal cancer. Irinotecan 90-100 ATP binding cassette subfamily B member 1 Homo sapiens 12-16 23801203-0 2013 [Correlation of MDR1 and ABCG2 genetic polymorphisms with the efficacy and adverse events of irinotecan chemotherapy in patients with colorectal cancer]. Irinotecan 93-103 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 25-30 23801203-1 2013 OBJECTIVE: To investigate the correlation of MDR1 and ABCG2 genetic polymorphisms with the efficacy and adverse events of irinotecan chemotherapy in patients with colorectal cancer (CRC). Irinotecan 122-132 ATP binding cassette subfamily B member 1 Homo sapiens 45-49 23801203-1 2013 OBJECTIVE: To investigate the correlation of MDR1 and ABCG2 genetic polymorphisms with the efficacy and adverse events of irinotecan chemotherapy in patients with colorectal cancer (CRC). Irinotecan 122-132 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 54-59 23377825-0 2013 Identification of cetrimonium bromide and irinotecan as compounds with synthetic lethality against NDRG1 deficient prostate cancer cells. Irinotecan 42-52 N-myc downstream regulated 1 Homo sapiens 99-104 23724141-7 2013 RESULTS: MSH3-deficient vs proficient CRC cells showed increased sensitivity to the irinotecan metabolite SN-38 and to oxaliplatin, but not 5-FU, as shown in assays for apoptosis and clonogenic survival. Irinotecan 84-94 mutS homolog 3 Homo sapiens 9-13 23724141-7 2013 RESULTS: MSH3-deficient vs proficient CRC cells showed increased sensitivity to the irinotecan metabolite SN-38 and to oxaliplatin, but not 5-FU, as shown in assays for apoptosis and clonogenic survival. Irinotecan 106-111 mutS homolog 3 Homo sapiens 9-13 23724141-9 2013 The impact of MSH3 knockdown on chemosensitivity to SN-38 and oxaliplatin was maintained independent of MLH1 status. Irinotecan 52-57 mutS homolog 3 Homo sapiens 14-18 23724141-10 2013 In MSH3-deficient vs proficient cells, SN-38 and oxaliplatin induced higher levels of phosphorylated histone H2AX and Chk2, and similar results were found in MLH1-proficient SW480 cells. Irinotecan 39-44 mutS homolog 3 Homo sapiens 3-7 23724141-10 2013 In MSH3-deficient vs proficient cells, SN-38 and oxaliplatin induced higher levels of phosphorylated histone H2AX and Chk2, and similar results were found in MLH1-proficient SW480 cells. Irinotecan 39-44 checkpoint kinase 2 Homo sapiens 118-122 23724141-10 2013 In MSH3-deficient vs proficient cells, SN-38 and oxaliplatin induced higher levels of phosphorylated histone H2AX and Chk2, and similar results were found in MLH1-proficient SW480 cells. Irinotecan 39-44 mutL homolog 1 Homo sapiens 158-162 23377825-6 2013 Screening of 3360 compounds revealed irinotecan and cetrimonium bromide (CTAB) as compounds that exhibited synthetic lethality against NDRG1 deficient PCa cells. Irinotecan 37-47 N-myc downstream regulated 1 Homo sapiens 135-140 23377825-9 2013 Our results suggest that CTAB and irinotecan could be further explored for their potential clinical benefit in patients with NDRG1 deficient PCa. Irinotecan 34-44 N-myc downstream regulated 1 Homo sapiens 125-130 24216984-3 2013 mTOR appears to enhance cancer cell survival following DNA damage, thus the inhibition of mTOR after irinotecan could theoretically show synergistic activities in patients. Irinotecan 101-111 mechanistic target of rapamycin kinase Homo sapiens 0-4 23439240-1 2013 Modified nanoprecipitation method was used for improved incorporation of hydrophilic drug (irinotecan hydrochloride) into the PLGA/PEO-PPO-PEO blended and blended/adsorbed nanoparticles. Irinotecan 91-115 protoporphyrinogen oxidase Homo sapiens 135-138 23461902-7 2013 RESULTS: We found that additional deficiency of Abcc4 in Abcb1a/b;Abcg2(-/-) mice significantly increased the brain concentration of all camptothecin analogues by 1.2-fold (gimatecan) to 5.8-fold (SN-38). Irinotecan 197-202 ATP-binding cassette, sub-family C (CFTR/MRP), member 4 Mus musculus 48-53 23461902-8 2013 The presence of Abcb1a/b or Abcc4 alone was sufficient to reduce the brain concentration of SN-38 to the level in WT mice. Irinotecan 92-97 ATP-binding cassette, sub-family B (MDR/TAP), member 1A Mus musculus 16-22 23461902-8 2013 The presence of Abcb1a/b or Abcc4 alone was sufficient to reduce the brain concentration of SN-38 to the level in WT mice. Irinotecan 92-97 ATP-binding cassette, sub-family C (CFTR/MRP), member 4 Mus musculus 28-33 24216984-3 2013 mTOR appears to enhance cancer cell survival following DNA damage, thus the inhibition of mTOR after irinotecan could theoretically show synergistic activities in patients. Irinotecan 101-111 mechanistic target of rapamycin kinase Homo sapiens 90-94 22124996-0 2013 Phase II study of bi-weekly irinotecan for patients with previously treated HER2-negative metastatic breast cancer: KMBOG0610B. Irinotecan 28-38 erb-b2 receptor tyrosine kinase 2 Homo sapiens 76-80 23564776-0 2013 ABCG2 inhibitor YHO-13351 sensitizes cancer stem/initiating-like side population cells to irinotecan. Irinotecan 90-100 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 0-5 23298313-7 2013 Second-line cetuximab plus irinotecan is a valid treatment strategy for mCRC patients with irinotecan-refractory and oxaliplatin-naive tumors harboring wild-type KRAS. Irinotecan 27-37 KRAS proto-oncogene, GTPase Homo sapiens 162-166 23358684-8 2013 Selection of irinotecan resistance in colon carcinoma cells led to a greater proportion of CSCs compared with parental cells, as measured by the CSC markers CD133 and ALDH1 activity (Aldefluor). Irinotecan 13-23 prominin 1 Homo sapiens 157-162 23373735-0 2013 Regulation of carboxylesterase-2 expression by p53 family proteins and enhanced anti-cancer activities among 5-fluorouracil, irinotecan and doxazolidine prodrug. Irinotecan 125-135 carboxylesterase 2 Homo sapiens 14-32 23373735-0 2013 Regulation of carboxylesterase-2 expression by p53 family proteins and enhanced anti-cancer activities among 5-fluorouracil, irinotecan and doxazolidine prodrug. Irinotecan 125-135 tumor protein p53 Homo sapiens 47-50 23373735-3 2013 Irinotecan and many others such as PPD (pentyl carbamate of p-aminobenzyl carbamate of doxazolidine) require activation by carboxylesterase-2 (CES2). Irinotecan 0-10 carboxylesterase 2 Homo sapiens 123-141 23373735-3 2013 Irinotecan and many others such as PPD (pentyl carbamate of p-aminobenzyl carbamate of doxazolidine) require activation by carboxylesterase-2 (CES2). Irinotecan 0-10 carboxylesterase 2 Homo sapiens 143-147 23358684-8 2013 Selection of irinotecan resistance in colon carcinoma cells led to a greater proportion of CSCs compared with parental cells, as measured by the CSC markers CD133 and ALDH1 activity (Aldefluor). Irinotecan 13-23 aldehyde dehydrogenase 1 family member A1 Homo sapiens 167-172 23770716-4 2013 Our data suggest that CPT-11 reduced the expression of IL-2/IFN-gamma and increased IL-10/TGF-beta expression in both peripheral blood and within the grafts. Irinotecan 22-28 interleukin 2 Mus musculus 55-59 23770716-4 2013 Our data suggest that CPT-11 reduced the expression of IL-2/IFN-gamma and increased IL-10/TGF-beta expression in both peripheral blood and within the grafts. Irinotecan 22-28 interferon gamma Mus musculus 60-69 23770716-4 2013 Our data suggest that CPT-11 reduced the expression of IL-2/IFN-gamma and increased IL-10/TGF-beta expression in both peripheral blood and within the grafts. Irinotecan 22-28 interleukin 10 Mus musculus 84-89 23770716-4 2013 Our data suggest that CPT-11 reduced the expression of IL-2/IFN-gamma and increased IL-10/TGF-beta expression in both peripheral blood and within the grafts. Irinotecan 22-28 transforming growth factor, beta 1 Mus musculus 90-98 23770716-5 2013 CPT-11 could also inhibit alloresponses of memory T cells, while decreasing the proportion of CD4(+) memory T cells in the spleen of the recipients and significantly reducing serum alloantibody levels. Irinotecan 0-6 CD4 antigen Mus musculus 94-97 23328075-3 2013 The aim of this study was to examine any modulations in the DDC mRNA levels in gastric cancer cells after their treatment with the chemotherapeutic agents 5-fluorouracil, leucovorin, irinotecan, etoposide, cisplatin, and taxol. Irinotecan 183-193 dopa decarboxylase Homo sapiens 60-63 23364789-6 2013 The PRC stress response was also activated by the topoisomerase I inhibitor 7-ethyl-10-hydroxycamptothecin (SN-38), an inducer of premature senescence in tumor cells. Irinotecan 76-106 PPARG related coactivator 1 Homo sapiens 4-7 23364789-6 2013 The PRC stress response was also activated by the topoisomerase I inhibitor 7-ethyl-10-hydroxycamptothecin (SN-38), an inducer of premature senescence in tumor cells. Irinotecan 108-113 PPARG related coactivator 1 Homo sapiens 4-7 23364789-7 2013 Cells treated with SN-38 displayed morphological characteristics of senescence and express senescence-associated beta-galactosidase activity. Irinotecan 19-24 galactosidase beta 1 Homo sapiens 113-131 23364789-8 2013 In contrast to menadione, the SN-38 induction of the PRC program occurred over an extended time course and was antioxidant-insensitive. Irinotecan 30-35 PPARG related coactivator 1 Homo sapiens 53-56 23547747-19 2013 The base-case incremental cost-effectiveness ratio (ICER) for KRAS WT patients for cetuximab compared with best supportive care is $98,000 per quality-adjusted life-year (QALY), for panitumumab compared with best supportive care is $150,000 per QALY and for cetuximab plus irinotecan compared with best supportive care is $88,000 per QALY. Irinotecan 273-283 KRAS proto-oncogene, GTPase Homo sapiens 62-66 23547747-22 2013 For cetuximab plus irinotecan treatment for KRAS WT people, there is no direct evidence on progression-free survival, overall survival and duration of treatment. Irinotecan 19-29 KRAS proto-oncogene, GTPase Homo sapiens 44-48 23547747-24 2013 It would be useful to conduct a RCT for patients with KRAS WT status receiving cetuximab plus irinotecan. Irinotecan 94-104 KRAS proto-oncogene, GTPase Homo sapiens 54-58 23054213-0 2013 The PARP inhibitor ABT-888 synergizes irinotecan treatment of colon cancer cell lines. Irinotecan 38-48 poly(ADP-ribose) polymerase 1 Homo sapiens 4-8 23386248-0 2013 Refining the UGT1A haplotype associated with irinotecan-induced hematological toxicity in metastatic colorectal cancer patients treated with 5-fluorouracil/irinotecan-based regimens. Irinotecan 45-55 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 13-18 23386248-0 2013 Refining the UGT1A haplotype associated with irinotecan-induced hematological toxicity in metastatic colorectal cancer patients treated with 5-fluorouracil/irinotecan-based regimens. Irinotecan 156-166 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 13-18 23386248-11 2013 Our results suggest that specific SNPs in UGT1A, other than UGT1A1*28, may influence irinotecan toxicity and should be considered to refine pharmacogenetic testing. Irinotecan 85-95 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 42-47 23985249-0 2013 [Relationship between UGT1A and irinotecan-related toxicity]. Irinotecan 32-42 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 22-27 23517622-5 2013 RESULTS: Gene expression levels of APTX, BRCA1 and ERCC1 were significantly lower in irinotecan-sensitive gastric cancer samples than those irinotecan-resistant samples (P<0.001 for all genes), while ISG15 (P=0.047) and Topo1 (P=0.002) were significantly higher. Irinotecan 85-95 aprataxin Mus musculus 35-39 23517622-5 2013 RESULTS: Gene expression levels of APTX, BRCA1 and ERCC1 were significantly lower in irinotecan-sensitive gastric cancer samples than those irinotecan-resistant samples (P<0.001 for all genes), while ISG15 (P=0.047) and Topo1 (P=0.002) were significantly higher. Irinotecan 85-95 breast cancer 1, early onset Mus musculus 41-46 23517622-5 2013 RESULTS: Gene expression levels of APTX, BRCA1 and ERCC1 were significantly lower in irinotecan-sensitive gastric cancer samples than those irinotecan-resistant samples (P<0.001 for all genes), while ISG15 (P=0.047) and Topo1 (P=0.002) were significantly higher. Irinotecan 85-95 excision repair cross-complementing rodent repair deficiency, complementation group 1 Mus musculus 51-56 23517622-5 2013 RESULTS: Gene expression levels of APTX, BRCA1 and ERCC1 were significantly lower in irinotecan-sensitive gastric cancer samples than those irinotecan-resistant samples (P<0.001 for all genes), while ISG15 (P=0.047) and Topo1 (P=0.002) were significantly higher. Irinotecan 85-95 ISG15 ubiquitin-like modifier Mus musculus 203-208 23517622-5 2013 RESULTS: Gene expression levels of APTX, BRCA1 and ERCC1 were significantly lower in irinotecan-sensitive gastric cancer samples than those irinotecan-resistant samples (P<0.001 for all genes), while ISG15 (P=0.047) and Topo1 (P=0.002) were significantly higher. Irinotecan 140-150 aprataxin Mus musculus 35-39 23517622-5 2013 RESULTS: Gene expression levels of APTX, BRCA1 and ERCC1 were significantly lower in irinotecan-sensitive gastric cancer samples than those irinotecan-resistant samples (P<0.001 for all genes), while ISG15 (P=0.047) and Topo1 (P=0.002) were significantly higher. Irinotecan 140-150 excision repair cross-complementing rodent repair deficiency, complementation group 1 Mus musculus 51-56 23480903-1 2013 BACKGROUND/PURPOSE: Human carboxylesterase 2 (CES2) is the key enzyme for metabolic activation of irinotecan (CPT-11). Irinotecan 98-108 carboxylesterase 2 Homo sapiens 26-44 23789755-0 2013 [Influence of genetic polymorphisms in UGT1A1, UGT1A7 and UGT1A9 on the pharmacokynetics of irinotecan, SN-38 and SN-38G]. Irinotecan 92-102 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 39-45 23789755-0 2013 [Influence of genetic polymorphisms in UGT1A1, UGT1A7 and UGT1A9 on the pharmacokynetics of irinotecan, SN-38 and SN-38G]. Irinotecan 92-102 UDP glucuronosyltransferase family 1 member A7 Homo sapiens 47-53 23789755-0 2013 [Influence of genetic polymorphisms in UGT1A1, UGT1A7 and UGT1A9 on the pharmacokynetics of irinotecan, SN-38 and SN-38G]. Irinotecan 92-102 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 58-64 23789755-0 2013 [Influence of genetic polymorphisms in UGT1A1, UGT1A7 and UGT1A9 on the pharmacokynetics of irinotecan, SN-38 and SN-38G]. Irinotecan 104-109 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 39-45 23789755-0 2013 [Influence of genetic polymorphisms in UGT1A1, UGT1A7 and UGT1A9 on the pharmacokynetics of irinotecan, SN-38 and SN-38G]. Irinotecan 104-109 UDP glucuronosyltransferase family 1 member A7 Homo sapiens 47-53 23789755-0 2013 [Influence of genetic polymorphisms in UGT1A1, UGT1A7 and UGT1A9 on the pharmacokynetics of irinotecan, SN-38 and SN-38G]. Irinotecan 104-109 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 58-64 23789755-0 2013 [Influence of genetic polymorphisms in UGT1A1, UGT1A7 and UGT1A9 on the pharmacokynetics of irinotecan, SN-38 and SN-38G]. Irinotecan 114-119 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 39-45 23789755-0 2013 [Influence of genetic polymorphisms in UGT1A1, UGT1A7 and UGT1A9 on the pharmacokynetics of irinotecan, SN-38 and SN-38G]. Irinotecan 114-119 UDP glucuronosyltransferase family 1 member A7 Homo sapiens 47-53 23789755-0 2013 [Influence of genetic polymorphisms in UGT1A1, UGT1A7 and UGT1A9 on the pharmacokynetics of irinotecan, SN-38 and SN-38G]. Irinotecan 114-119 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 58-64 23789755-1 2013 OBJECTIVE: To evaluate the influence of genetic polymorphism in UGT1A1, UGT1A7 and UGT1A9 on the population pharmacokinetics of irinotecan and its metabolites, SN-38 and SN-38G. Irinotecan 128-138 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 64-70 23789755-1 2013 OBJECTIVE: To evaluate the influence of genetic polymorphism in UGT1A1, UGT1A7 and UGT1A9 on the population pharmacokinetics of irinotecan and its metabolites, SN-38 and SN-38G. Irinotecan 128-138 UDP glucuronosyltransferase family 1 member A7 Homo sapiens 72-78 23789755-1 2013 OBJECTIVE: To evaluate the influence of genetic polymorphism in UGT1A1, UGT1A7 and UGT1A9 on the population pharmacokinetics of irinotecan and its metabolites, SN-38 and SN-38G. Irinotecan 128-138 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 83-89 23789755-7 2013 The presence of UGT1A1*28, UGT1A7*3, UGT1A9*22 genotypes decreases SN-38 clearance between 20 and 36%. Irinotecan 67-72 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 16-22 23789755-7 2013 The presence of UGT1A1*28, UGT1A7*3, UGT1A9*22 genotypes decreases SN-38 clearance between 20 and 36%. Irinotecan 67-72 UDP glucuronosyltransferase family 1 member A7 Homo sapiens 27-33 23789755-7 2013 The presence of UGT1A1*28, UGT1A7*3, UGT1A9*22 genotypes decreases SN-38 clearance between 20 and 36%. Irinotecan 67-72 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 37-43 23789755-9 2013 CONCLUSION: The inclusion of pharmacokinetic-pharmacogenomic information can add value to the individualized dose adjustment of irinotecan, because it will let quantitatively handle dose reductions in patients with iatrogenic toxicity due to UGT1A1 genetic polymorphisms. Irinotecan 128-138 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 242-248 23480903-1 2013 BACKGROUND/PURPOSE: Human carboxylesterase 2 (CES2) is the key enzyme for metabolic activation of irinotecan (CPT-11). Irinotecan 98-108 carboxylesterase 2 Homo sapiens 46-50 23480903-1 2013 BACKGROUND/PURPOSE: Human carboxylesterase 2 (CES2) is the key enzyme for metabolic activation of irinotecan (CPT-11). Irinotecan 110-116 carboxylesterase 2 Homo sapiens 26-44 23480903-1 2013 BACKGROUND/PURPOSE: Human carboxylesterase 2 (CES2) is the key enzyme for metabolic activation of irinotecan (CPT-11). Irinotecan 110-116 carboxylesterase 2 Homo sapiens 46-50 23480903-8 2013 RESULTS: CES2 RNA expression was observed in neuroblastoma cells, and its expression in neuroblastoma cell lines was positively correlated with sensitivity to CPT-11 and apoptosis after CPT-11 exposure in vitro. Irinotecan 159-165 carboxylesterase 2 Homo sapiens 9-13 23480903-8 2013 RESULTS: CES2 RNA expression was observed in neuroblastoma cells, and its expression in neuroblastoma cell lines was positively correlated with sensitivity to CPT-11 and apoptosis after CPT-11 exposure in vitro. Irinotecan 186-192 carboxylesterase 2 Homo sapiens 9-13 23041588-1 2013 BACKGROUND: The purpose of this study was to evaluate the combination of panitumumab and irinotecan in patients with KRAS wild-type metastatic colorectal cancer refractory to standard chemotherapy (oxaliplatin, fluoropyrimidines-irinotecan and bevacizumab). Irinotecan 89-99 KRAS proto-oncogene, GTPase Homo sapiens 117-121 23041588-1 2013 BACKGROUND: The purpose of this study was to evaluate the combination of panitumumab and irinotecan in patients with KRAS wild-type metastatic colorectal cancer refractory to standard chemotherapy (oxaliplatin, fluoropyrimidines-irinotecan and bevacizumab). Irinotecan 229-239 KRAS proto-oncogene, GTPase Homo sapiens 117-121 23130636-2 2013 UGT1A1 (UDP-glucuronosyltransferase 1A1) is a critical gene for bilirubin metabolism and irinotecan detoxification. Irinotecan 89-99 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 23108760-0 2013 Fatty acid synthase inhibitor cerulenin inhibits topoisomerase I catalytic activity and augments SN-38-induced apoptosis. Irinotecan 97-102 fatty acid synthase Homo sapiens 0-19 22934695-2 2013 This study aims to investigate the association between UGT1A1 gene polymorphisms and irinotecan related toxicities in Chinese Han gastrointestinal cancer patients receiving FOLFIRI regimen chemotherapy. Irinotecan 85-95 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 55-61 23130636-2 2013 UGT1A1 (UDP-glucuronosyltransferase 1A1) is a critical gene for bilirubin metabolism and irinotecan detoxification. Irinotecan 89-99 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 8-39 22402322-5 2013 Here, we have used an established, genetically modified human NSC line (HB1.F3.CD) to deliver carboxylesterase (CE) to cerebellar tumor foci and locally activate the prodrug camptothecin-11 (CPT-11) (Irinotecan) to the potent topoisomerase I inhibitor SN-38. Irinotecan 174-189 histocompatibility minor HB-1 Homo sapiens 72-75 23121194-5 2013 On the other hand, anti-CD 20 mAb-PEG-SN-38 via carbamate-bond as conventional immunoconjugate, enabled SN-38 to be released by a carboxylesterase inside of the tumor cell following the internalization, showed strong anti-tumor activity against malignant lymphoma as hypervascular and stroma-poor tumor. Irinotecan 38-43 keratin 20 Homo sapiens 24-29 23348635-6 2013 Nonetheless, cells exposed to 5-FU, CPT-11, TNF-alpha or HSV-1 activate NF-kappaB. Irinotecan 36-42 nuclear factor kappa B subunit 1 Homo sapiens 72-81 23121194-5 2013 On the other hand, anti-CD 20 mAb-PEG-SN-38 via carbamate-bond as conventional immunoconjugate, enabled SN-38 to be released by a carboxylesterase inside of the tumor cell following the internalization, showed strong anti-tumor activity against malignant lymphoma as hypervascular and stroma-poor tumor. Irinotecan 104-109 keratin 20 Homo sapiens 24-29 22402322-5 2013 Here, we have used an established, genetically modified human NSC line (HB1.F3.CD) to deliver carboxylesterase (CE) to cerebellar tumor foci and locally activate the prodrug camptothecin-11 (CPT-11) (Irinotecan) to the potent topoisomerase I inhibitor SN-38. Irinotecan 191-197 histocompatibility minor HB-1 Homo sapiens 72-75 22402322-5 2013 Here, we have used an established, genetically modified human NSC line (HB1.F3.CD) to deliver carboxylesterase (CE) to cerebellar tumor foci and locally activate the prodrug camptothecin-11 (CPT-11) (Irinotecan) to the potent topoisomerase I inhibitor SN-38. Irinotecan 200-210 histocompatibility minor HB-1 Homo sapiens 72-75 22402322-5 2013 Here, we have used an established, genetically modified human NSC line (HB1.F3.CD) to deliver carboxylesterase (CE) to cerebellar tumor foci and locally activate the prodrug camptothecin-11 (CPT-11) (Irinotecan) to the potent topoisomerase I inhibitor SN-38. Irinotecan 252-257 histocompatibility minor HB-1 Homo sapiens 72-75 23449330-5 2013 Then the kinetics of estradiol at 3-hydroxy position (E-3OH) and 7-ethyl-10-hydroxycamptothecin (SN-38) glucuronidation by recombinant UGT1A1s as well as human and minipig liver microsomes were analyzed. Irinotecan 65-95 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 135-141 23278696-5 2013 KEY FINDINGS: Genistein, as well as probenecid, significantly suppressed the MRP2-mediated biliary and intestinal secretion of CPT-11 and its metabolites and increased their plasma concentrations. Irinotecan 127-133 ATP binding cassette subfamily C member 2 Rattus norvegicus 77-81 23303296-4 2013 Since SN-38 is metabolized to non-toxic SN-38-glucuronide by hepatic uridine diphosphate glucuronosyl transferase (UGT) 1A enzymes, UGT1A enzyme activities may influence adverse events of CPT-11. Irinotecan 6-11 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 115-122 23303296-4 2013 Since SN-38 is metabolized to non-toxic SN-38-glucuronide by hepatic uridine diphosphate glucuronosyl transferase (UGT) 1A enzymes, UGT1A enzyme activities may influence adverse events of CPT-11. Irinotecan 6-11 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 132-137 23303296-4 2013 Since SN-38 is metabolized to non-toxic SN-38-glucuronide by hepatic uridine diphosphate glucuronosyl transferase (UGT) 1A enzymes, UGT1A enzyme activities may influence adverse events of CPT-11. Irinotecan 188-194 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 115-122 23303296-4 2013 Since SN-38 is metabolized to non-toxic SN-38-glucuronide by hepatic uridine diphosphate glucuronosyl transferase (UGT) 1A enzymes, UGT1A enzyme activities may influence adverse events of CPT-11. Irinotecan 188-194 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 132-137 23303296-6 2013 Here, to identify the genetic factors that affect the adverse events of CPT-11, we determined the polymorphism in three UGT 1A isozyme genes in 45 inpatients with lung, colon, or stomach cancer. Irinotecan 72-78 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 120-126 23303296-12 2013 In conclusion, UGT1A1*6/*28, UGT1A7*3/*3, and UGT1A9*1/*1 should be taken into consideration as markers for preventing severe adverse events of CPT-11 administration. Irinotecan 144-150 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 15-21 23303296-12 2013 In conclusion, UGT1A1*6/*28, UGT1A7*3/*3, and UGT1A9*1/*1 should be taken into consideration as markers for preventing severe adverse events of CPT-11 administration. Irinotecan 144-150 UDP glucuronosyltransferase family 1 member A7 Homo sapiens 29-35 23303296-12 2013 In conclusion, UGT1A1*6/*28, UGT1A7*3/*3, and UGT1A9*1/*1 should be taken into consideration as markers for preventing severe adverse events of CPT-11 administration. Irinotecan 144-150 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 46-52 23089672-11 2013 Regarding phase II enzyme polymorphism, the anticancer treatment with mercaptopurines and irinotecan is still considered important in relation to the polymorphism of TPMT and UGT1A1, respectively. Irinotecan 90-100 thiopurine S-methyltransferase Homo sapiens 166-170 23089672-11 2013 Regarding phase II enzyme polymorphism, the anticancer treatment with mercaptopurines and irinotecan is still considered important in relation to the polymorphism of TPMT and UGT1A1, respectively. Irinotecan 90-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 175-181 23449330-7 2013 E-3OH and SN-38 glucuronidation by recombinant hUGT1A1 and mpUGT1A1 showed allosteric sigmoidal kinetics. Irinotecan 10-15 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 47-54 23449330-5 2013 Then the kinetics of estradiol at 3-hydroxy position (E-3OH) and 7-ethyl-10-hydroxycamptothecin (SN-38) glucuronidation by recombinant UGT1A1s as well as human and minipig liver microsomes were analyzed. Irinotecan 97-102 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 135-141 22973964-0 2013 Irinotecan resistance is accompanied by upregulation of EGFR and Src signaling in human cancer models. Irinotecan 0-10 epidermal growth factor receptor Homo sapiens 56-60 22973964-6 2013 We now show that acquired resistance to SN-38 is accompanied by increased expression of EGFR, HER2, HER3 and Src proteins in two colorectal cancer cell models as well as by Src activation. Irinotecan 40-45 erb-b2 receptor tyrosine kinase 2 Homo sapiens 94-98 23242585-13 2013 Immunostains of gammaH2AX and pChk1(S296) on serial tumor biopsies from four patients demonstrated an induction of DNA damage and Chk1 activation following irinotecan. Irinotecan 156-166 checkpoint kinase 1 Homo sapiens 31-35 23053265-0 2013 Uridine diphosphate glucuronosyl transferase 1 family polypeptide A1 gene (UGT1A1) polymorphisms are associated with toxicity and efficacy in irinotecan monotherapy for refractory pancreatic cancer. Irinotecan 142-152 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 75-81 24457609-0 2013 Gefitinib enhances the antitumor activity of CPT-11 in vitro and in vivo by inhibiting ABCG2 but not ABCB1: a new clue to circumvent gastrointestinal toxicity risk. Irinotecan 45-51 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 87-92 24457609-2 2013 METHODS: We aimed to evaluate the inhibitory potency of gefitinib against the ABCB1- or ABCG2-mediated excretion of CPT-11 and its active metabolite SN-38 in vitro and in vivo. Irinotecan 116-122 ATP binding cassette subfamily B member 1 Homo sapiens 78-83 24457609-2 2013 METHODS: We aimed to evaluate the inhibitory potency of gefitinib against the ABCB1- or ABCG2-mediated excretion of CPT-11 and its active metabolite SN-38 in vitro and in vivo. Irinotecan 116-122 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 88-93 24457609-2 2013 METHODS: We aimed to evaluate the inhibitory potency of gefitinib against the ABCB1- or ABCG2-mediated excretion of CPT-11 and its active metabolite SN-38 in vitro and in vivo. Irinotecan 149-154 ATP binding cassette subfamily B member 1 Homo sapiens 78-83 24457609-2 2013 METHODS: We aimed to evaluate the inhibitory potency of gefitinib against the ABCB1- or ABCG2-mediated excretion of CPT-11 and its active metabolite SN-38 in vitro and in vivo. Irinotecan 149-154 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 88-93 24457609-3 2013 RESULTS: Gefitinib dose-dependently enhanced the antiproliferation activity of SN-38 in vitro by inhibiting ABCG2. Irinotecan 79-84 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 108-113 22973964-6 2013 We now show that acquired resistance to SN-38 is accompanied by increased expression of EGFR, HER2, HER3 and Src proteins in two colorectal cancer cell models as well as by Src activation. Irinotecan 40-45 erb-b2 receptor tyrosine kinase 3 Homo sapiens 100-104 22973964-6 2013 We now show that acquired resistance to SN-38 is accompanied by increased expression of EGFR, HER2, HER3 and Src proteins in two colorectal cancer cell models as well as by Src activation. Irinotecan 40-45 SRC proto-oncogene, non-receptor tyrosine kinase Homo sapiens 109-112 22973964-6 2013 We now show that acquired resistance to SN-38 is accompanied by increased expression of EGFR, HER2, HER3 and Src proteins in two colorectal cancer cell models as well as by Src activation. Irinotecan 40-45 SRC proto-oncogene, non-receptor tyrosine kinase Homo sapiens 173-176 22973964-0 2013 Irinotecan resistance is accompanied by upregulation of EGFR and Src signaling in human cancer models. Irinotecan 0-10 SRC proto-oncogene, non-receptor tyrosine kinase Homo sapiens 65-68 22973964-4 2013 Exposure of tumor cells to irinotecan or its active metabolite SN-38 is accompanied by EGFR activation, either by stimulation of EGFR autophosphorylation or by Src-mediated phosphorylation. Irinotecan 27-37 epidermal growth factor receptor Homo sapiens 87-91 22973964-4 2013 Exposure of tumor cells to irinotecan or its active metabolite SN-38 is accompanied by EGFR activation, either by stimulation of EGFR autophosphorylation or by Src-mediated phosphorylation. Irinotecan 27-37 epidermal growth factor receptor Homo sapiens 129-133 22973964-4 2013 Exposure of tumor cells to irinotecan or its active metabolite SN-38 is accompanied by EGFR activation, either by stimulation of EGFR autophosphorylation or by Src-mediated phosphorylation. Irinotecan 27-37 SRC proto-oncogene, non-receptor tyrosine kinase Homo sapiens 160-163 22973964-4 2013 Exposure of tumor cells to irinotecan or its active metabolite SN-38 is accompanied by EGFR activation, either by stimulation of EGFR autophosphorylation or by Src-mediated phosphorylation. Irinotecan 63-68 epidermal growth factor receptor Homo sapiens 87-91 22973964-4 2013 Exposure of tumor cells to irinotecan or its active metabolite SN-38 is accompanied by EGFR activation, either by stimulation of EGFR autophosphorylation or by Src-mediated phosphorylation. Irinotecan 63-68 epidermal growth factor receptor Homo sapiens 129-133 22973964-4 2013 Exposure of tumor cells to irinotecan or its active metabolite SN-38 is accompanied by EGFR activation, either by stimulation of EGFR autophosphorylation or by Src-mediated phosphorylation. Irinotecan 63-68 SRC proto-oncogene, non-receptor tyrosine kinase Homo sapiens 160-163 22973964-6 2013 We now show that acquired resistance to SN-38 is accompanied by increased expression of EGFR, HER2, HER3 and Src proteins in two colorectal cancer cell models as well as by Src activation. Irinotecan 40-45 epidermal growth factor receptor Homo sapiens 88-92 23783485-0 2013 Associations between UGT1A1*6/*28 polymorphisms and irinotecan-induced severe toxicity in Chinese gastric or esophageal cancer patients. Irinotecan 52-62 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 21-27 23089802-3 2013 Impaired or reduced UGT1A1 activity causes unconjugated hyperbilirubinemia (Gilbert"s syndrome and Crigler-Najjar syndrome) and side effects of drug treatment such as SN-38 (active metabolite of the anticancer drug irinotecan)-induced toxicity. Irinotecan 167-172 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 20-26 23781580-0 2013 UGT1A1 genotype-specific phase I and pharmacokinetic study for combination chemotherapy with irinotecan and cisplatin: a Saitama Tumor Board study. Irinotecan 93-103 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 23781580-2 2013 The aim of the present study was to determine the RD of irinotecan in combination with cisplatin (CPT-P) for individuals with or without UGT1A1 polymorphisms. Irinotecan 56-66 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 137-143 23781580-8 2013 CONCLUSION: UGT1A1 genotype-based RDs of irinotecan in CPT-P therapy were determined. Irinotecan 41-51 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-18 23316441-3 2013 In this study, we focused on camptothecin and its derivatives, topotecan and irinotecan, which are type I topoisomerase inhibitors that create DNA double-strand breaks in rapidly dividing cells. Irinotecan 77-87 Topoisomerase 1 Drosophila melanogaster 99-119 23686699-0 2013 UGT1A1 6/28 polymorphisms could predict irinotecan-induced severe neutropenia not diarrhea in Chinese colorectal cancer patients. Irinotecan 40-50 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 23686699-1 2013 The aim of this study was to investigate the associations between UDP-glucuronosyltransferase (UGT) 1A1 polymorphisms and irinotecan-induced toxicities in Chinese advanced colorectal cancer patients. Irinotecan 122-132 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 66-103 23686699-9 2013 Both UGT1A1 6 and UGT1A1 28 variants were closely associated with irinotecan-induced severe neutropenia, but not diarrhea. Irinotecan 66-76 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 5-11 23686699-9 2013 Both UGT1A1 6 and UGT1A1 28 variants were closely associated with irinotecan-induced severe neutropenia, but not diarrhea. Irinotecan 66-76 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 18-24 23089802-3 2013 Impaired or reduced UGT1A1 activity causes unconjugated hyperbilirubinemia (Gilbert"s syndrome and Crigler-Najjar syndrome) and side effects of drug treatment such as SN-38 (active metabolite of the anticancer drug irinotecan)-induced toxicity. Irinotecan 215-225 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 20-26 23648675-1 2013 Human carboxylesterase (CES) 1A and CES2, two major forms of human CES, dominate the pharmacokinetics of most prodrugs such as imidapril and irinotecan (CPT-11). Irinotecan 141-151 carboxylesterase 2 Homo sapiens 36-40 23648675-1 2013 Human carboxylesterase (CES) 1A and CES2, two major forms of human CES, dominate the pharmacokinetics of most prodrugs such as imidapril and irinotecan (CPT-11). Irinotecan 153-159 carboxylesterase 2 Homo sapiens 36-40 23783485-1 2013 The aim of this study was to investigate the associations between UDP-glucuronosyltransferase (UGT) 1A1 polymorphisms and irinotecan-induced toxicities in Chinese advanced gastric or esophageal cancer patients. Irinotecan 122-132 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 66-103 23864251-2 2013 Therefore, we conducted this study to evaluate the effect of the combination of irinotecan and cisplatin in the treatment of GEP-NECs. Irinotecan 80-90 granulin precursor Homo sapiens 125-128 23864251-13 2013 The irinotecan plus cisplatin chemotherapy is moderately effective and tolerable well tolerated in advanced or metastatic GEP-NEC patients; octreotide LAR may be a good maintenance treatment and should be considered as a treatment option for these patients in the future. Irinotecan 4-14 granulin precursor Homo sapiens 122-125 23555046-9 2013 siRNA-mediated silencing of EGFR and Rictor reduced U251MG cell migration and increased sensitivity of the cells to irinotecan, temozolomide and vincristine. Irinotecan 116-126 epidermal growth factor receptor Homo sapiens 28-32 23422270-0 2013 The role of p38 in irinotecan-induced DNA damage and apoptosis of colon cancer cells. Irinotecan 19-29 mitogen-activated protein kinase 14 Homo sapiens 12-15 23422270-1 2013 The role of p38 in irinotecan (CPT-11)-induced damage and cell death in colon cancer cell line SW620 was investigated. Irinotecan 19-29 mitogen-activated protein kinase 14 Homo sapiens 12-15 23422270-2 2013 We demonstrate that CPT-11 treatment activates p38 in exposed cells, however with concentration dependent dynamics and differing consequences. Irinotecan 20-26 mitogen-activated protein kinase 14 Homo sapiens 47-50 23422270-3 2013 Higher CPT-11 concentrations induce a massive early but relatively short-lasting p38 activity leading to apoptosis mediated by mitochondria and caspases. Irinotecan 7-13 mitogen-activated protein kinase 14 Homo sapiens 81-84 23422270-4 2013 Pharmacological or siRNA inhibition of p38 then significantly prevents CPT-11-dependent cell death. Irinotecan 71-77 mitogen-activated protein kinase 14 Homo sapiens 39-42 23422270-5 2013 Conversely, lower CPT-11 concentrations activate p38 in a delayed, however sustained manner, with apoptosis occurring only in a fraction of cells and in the absence of significant autophagy. Irinotecan 18-24 mitogen-activated protein kinase 14 Homo sapiens 49-52 23422270-6 2013 Blocking p38 in thus treated cells increases their sensitivity toward CPT-11 and increases cell death. Irinotecan 70-76 mitogen-activated protein kinase 14 Homo sapiens 9-12 23422270-7 2013 In summary, our results confirm the involvement of p38 in colon cancer cells response to CPT-11 while indicating a varying role of p38 in the final biological response. Irinotecan 89-95 mitogen-activated protein kinase 14 Homo sapiens 51-54 23328486-0 2013 A phase II study of high-dose cetuximab plus irinotecan in colorectal cancer patients with KRAS wild-type tumors who progressed after standard dose of cetuximab plus irinotecan. Irinotecan 45-55 KRAS proto-oncogene, GTPase Homo sapiens 91-95 23328486-1 2013 OBJECTIVES: We conducted a phase II clinical trial of high-dose cetuximab plus irinotecan in KRAS wild-type patients who progressed on standard-dose cetuximab plus irinotecan. Irinotecan 79-89 KRAS proto-oncogene, GTPase Homo sapiens 93-97 23555046-9 2013 siRNA-mediated silencing of EGFR and Rictor reduced U251MG cell migration and increased sensitivity of the cells to irinotecan, temozolomide and vincristine. Irinotecan 116-126 RPTOR independent companion of MTOR complex 2 Homo sapiens 37-43 23516488-0 2013 Association between UGT1A1*28 polymorphisms and clinical outcomes of irinotecan-based chemotherapies in colorectal cancer: a meta-analysis in Caucasians. Irinotecan 69-79 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 20-26 23516488-1 2013 BACKGROUND: Whether UGT1A1*28 genotype is associated with clinical outcomes of irinotecan (IRI)-based chemotherapy in Colorectal cancer (CRC) is an important gap in existing knowledge to inform clinical utility. Irinotecan 79-89 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 20-26 24292186-4 2013 UGT1A1*6 and *28 were significantly associated with altered pharmacokinetics of an irinotecan metabolite, SN-38, and with increased frequency of severe neutropenia. Irinotecan 83-93 UDP glucuronosyltransferase 1 family, polypeptide A1 Mus musculus 0-6 23520486-2 2013 As such, we sought to determine the ability of the mTOR inhibitor everolimus to potentiate the antitumor effects of irinotecan in colorectal cancer (CRC). Irinotecan 116-126 mechanistic target of rapamycin kinase Mus musculus 51-55 23520486-11 2013 CONCLUSIONS: Quantitative analysis of tumor growth and metabolomic data showed that the combination of everolimus and irinotecan was more beneficial in the BRAF/PIK3CA mutant HT29 tumor xenografts, which had an additive effect, than the KRAS/PIK3CA mutant HCT116 tumor xenografts, which had a less than additive effect. Irinotecan 118-128 Braf transforming gene Mus musculus 156-160 23520486-11 2013 CONCLUSIONS: Quantitative analysis of tumor growth and metabolomic data showed that the combination of everolimus and irinotecan was more beneficial in the BRAF/PIK3CA mutant HT29 tumor xenografts, which had an additive effect, than the KRAS/PIK3CA mutant HCT116 tumor xenografts, which had a less than additive effect. Irinotecan 118-128 phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha Mus musculus 161-167 23520486-11 2013 CONCLUSIONS: Quantitative analysis of tumor growth and metabolomic data showed that the combination of everolimus and irinotecan was more beneficial in the BRAF/PIK3CA mutant HT29 tumor xenografts, which had an additive effect, than the KRAS/PIK3CA mutant HCT116 tumor xenografts, which had a less than additive effect. Irinotecan 118-128 Kirsten rat sarcoma viral oncogene homolog Mus musculus 237-241 23520486-11 2013 CONCLUSIONS: Quantitative analysis of tumor growth and metabolomic data showed that the combination of everolimus and irinotecan was more beneficial in the BRAF/PIK3CA mutant HT29 tumor xenografts, which had an additive effect, than the KRAS/PIK3CA mutant HCT116 tumor xenografts, which had a less than additive effect. Irinotecan 118-128 phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha Mus musculus 242-248 24036898-6 2013 Although SN-38-resistant SCLC cells lacking ICAM-1 expression were still refractory to trastuzumab, their in vivo growth was significantly suppressed by bevacizumab treatment due to dependence on their distinctive and abundant production of vascular endothelial growth factor. Irinotecan 9-14 intercellular adhesion molecule 1 Homo sapiens 44-50 24036898-6 2013 Although SN-38-resistant SCLC cells lacking ICAM-1 expression were still refractory to trastuzumab, their in vivo growth was significantly suppressed by bevacizumab treatment due to dependence on their distinctive and abundant production of vascular endothelial growth factor. Irinotecan 9-14 vascular endothelial growth factor A Homo sapiens 241-275 23328499-6 2013 SN-38 glucuronidation in human and rat liver microsomes was inhibited dose-dependently by the presence of tacrolimus and the 50% inhibition concentration (IC50) values of tacrolimus in rat and human liver microsomes were 10.33 muM and 3.58 muM, respectively. Irinotecan 0-5 latexin Homo sapiens 227-230 23328499-6 2013 SN-38 glucuronidation in human and rat liver microsomes was inhibited dose-dependently by the presence of tacrolimus and the 50% inhibition concentration (IC50) values of tacrolimus in rat and human liver microsomes were 10.33 muM and 3.58 muM, respectively. Irinotecan 0-5 latexin Homo sapiens 240-243 23328499-8 2013 These findings suggest that tacrolimus inhibits UGT1A1-mediated SN-38 glucuronidation. Irinotecan 64-69 UDP glucuronosyltransferase family 1 member A1 Rattus norvegicus 48-54 23811769-8 2013 We also attempted to ameliorate the intestinal toxicity of irinotecan hydrochloride by modulating the hepatic and intestinal functions of MRP2. Irinotecan 59-83 ATP binding cassette subfamily C member 2 Homo sapiens 138-142 24292186-4 2013 UGT1A1*6 and *28 were significantly associated with altered pharmacokinetics of an irinotecan metabolite, SN-38, and with increased frequency of severe neutropenia. Irinotecan 106-111 UDP glucuronosyltransferase 1 family, polypeptide A1 Mus musculus 0-6 23236239-0 2012 UGT1A1 predicts outcome in colorectal cancer treated with irinotecan and fluorouracil. Irinotecan 58-68 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 23149859-10 2012 After SN-38 was released from TPGS/PLGA/SN-38 NPs in MDR cells, TPGS or/and PLGA may modulate the efflux microenvironment of the P-gp pump, such as mitochondria and the P-gp domain with an ATP-binding site. Irinotecan 6-11 ATP binding cassette subfamily B member 1 Homo sapiens 129-133 23149859-10 2012 After SN-38 was released from TPGS/PLGA/SN-38 NPs in MDR cells, TPGS or/and PLGA may modulate the efflux microenvironment of the P-gp pump, such as mitochondria and the P-gp domain with an ATP-binding site. Irinotecan 6-11 ATP binding cassette subfamily B member 1 Homo sapiens 169-173 23149859-10 2012 After SN-38 was released from TPGS/PLGA/SN-38 NPs in MDR cells, TPGS or/and PLGA may modulate the efflux microenvironment of the P-gp pump, such as mitochondria and the P-gp domain with an ATP-binding site. Irinotecan 40-45 ATP binding cassette subfamily B member 1 Homo sapiens 129-133 23149859-10 2012 After SN-38 was released from TPGS/PLGA/SN-38 NPs in MDR cells, TPGS or/and PLGA may modulate the efflux microenvironment of the P-gp pump, such as mitochondria and the P-gp domain with an ATP-binding site. Irinotecan 40-45 ATP binding cassette subfamily B member 1 Homo sapiens 169-173 20301284-8 1993 Agents/circumstances to avoid: Because TDP1 codes for a DNA repair enzyme, genotoxic anti-cancer drugs such as camptothecins (e.g., irinotecan and topotecan) and bleomycin are likely to be extremely harmful and possibly fatal; exposure to radiation is likely to be extremely harmful and possibly fatal. Irinotecan 132-142 tyrosyl-DNA phosphodiesterase 1 Homo sapiens 39-43 23236239-9 2012 Differences in irinotecan and its metabolites between UGT1A1 gene variants were compared. Irinotecan 15-25 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 54-60 23236239-21 2012 Irinotecan PK was investigated in 34 patients, which showed high area under concentration curve (AUC) of irinotecan and SN-38, but low AUC ratio (SN-38G / SN-38) in those patients with UGT1A1*28 TA7/7. Irinotecan 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 185-191 23236239-22 2012 CONCLUSION: A distinct distribution pattern of UGT1A1 genotypes in Chinese patients might contribute to relatively low toxicity associated with irinotecan and 5-fluorouracil in mCRC patients. Irinotecan 144-154 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 47-53 23225456-7 2012 CONCLUSION: Our data demonstrate the efficacy of HAI of irinotecan combined with 5-fluorouracil and leucovorin for liver metastases from CRC, specifically in patients also treated with LDP. Irinotecan 56-66 carboxypeptidase Q Homo sapiens 185-188 23081779-6 2012 Upon exposure to irinotecan, the LGR5(+) cells transitioned into an LGR5(-) drug-resistant state. Irinotecan 17-27 leucine rich repeat containing G protein-coupled receptor 5 Homo sapiens 33-37 23071293-0 2012 Clinical outcome of Japanese metastatic colorectal cancer patients harbouring the KRAS p.G13D mutation treated with cetuximab + irinotecan. Irinotecan 128-138 KRAS proto-oncogene, GTPase Homo sapiens 82-86 21844885-0 2012 Vascular endothelial growth factor polymorphisms and clinical outcome in colorectal cancer patients treated with irinotecan-based chemotherapy and bevacizumab. Irinotecan 113-123 vascular endothelial growth factor A Homo sapiens 0-34 21844885-10 2012 Our results support the potential predictive ability of VEGF genotypes in patients with metastatic colorectal cancer receiving irinotecan-based chemotherapy plus bevacizumab, in terms of RR and OS. Irinotecan 127-137 vascular endothelial growth factor A Homo sapiens 56-60 22753594-10 2012 We found that DPC4/SMAD4 inactivation sensitized pancreatic cancer cells to cisplatin and irinotecan by 2- to 4-fold, but they were modestly less sensitive to gemcitabine. Irinotecan 90-100 SMAD family member 4 Homo sapiens 14-18 22753594-10 2012 We found that DPC4/SMAD4 inactivation sensitized pancreatic cancer cells to cisplatin and irinotecan by 2- to 4-fold, but they were modestly less sensitive to gemcitabine. Irinotecan 90-100 SMAD family member 4 Homo sapiens 19-24 22270257-10 2012 Conversely potency of PP242 and the combination index for PP242 + irinotecan were unrelated, but synergy exists across all dose levels in PP242 and irinotecan sensitive, p53 wild-type cell lines. Irinotecan 148-158 tumor protein p53 Homo sapiens 170-173 23271276-3 2012 Imidapril and irinotecan hydrochloride (CPT-11) were used as special substrates for CES1 and CES2, respectively. Irinotecan 14-38 carboxylesterase 1E Rattus norvegicus 84-88 23271276-3 2012 Imidapril and irinotecan hydrochloride (CPT-11) were used as special substrates for CES1 and CES2, respectively. Irinotecan 14-38 carboxylesterase 2 Rattus norvegicus 93-97 23271276-3 2012 Imidapril and irinotecan hydrochloride (CPT-11) were used as special substrates for CES1 and CES2, respectively. Irinotecan 40-46 carboxylesterase 1E Rattus norvegicus 84-88 23271276-3 2012 Imidapril and irinotecan hydrochloride (CPT-11) were used as special substrates for CES1 and CES2, respectively. Irinotecan 40-46 carboxylesterase 2 Rattus norvegicus 93-97 23081779-6 2012 Upon exposure to irinotecan, the LGR5(+) cells transitioned into an LGR5(-) drug-resistant state. Irinotecan 17-27 leucine rich repeat containing G protein-coupled receptor 5 Homo sapiens 68-72 23233861-7 2012 Of the proposed gene/drug pairs the highest importance was assigned to HLA-B/abacavir, UGT1A1/irinotecan, and CYP2D6/tamoxifen. Irinotecan 94-104 major histocompatibility complex, class I, B Homo sapiens 71-76 23233861-7 2012 Of the proposed gene/drug pairs the highest importance was assigned to HLA-B/abacavir, UGT1A1/irinotecan, and CYP2D6/tamoxifen. Irinotecan 94-104 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 87-93 23155248-5 2012 SN-38 induced different types of cell death in two OSCC cell lines: apoptosis (caspase-3 activation and internucleosomal DNA fragmentation) in HSC-2 cells and autophagy (formation of autophagosome and secondary lysosome) in HSC-4 cells. Irinotecan 0-5 caspase 3 Homo sapiens 79-88 23162643-1 2012 This study retrospectively compared nedaplatin and irinotecan hydrochloride (NDP/CPT) combination therapy with cisplatin and irinotecan hydrochloride therapy (CDDP/CPT) for efficacy and adverse events in the treatment of clear cell adenocarcinoma of the ovary (CCC) and recurrent ovarian carcinoma. Irinotecan 51-75 norrin cystine knot growth factor NDP Homo sapiens 77-80 22909976-0 2012 K-Ras gene mutation status as a prognostic and predictive factor in patients with colorectal cancer undergoing irinotecan- or oxaliplatin-based chemotherapy. Irinotecan 111-121 KRAS proto-oncogene, GTPase Homo sapiens 0-5 22909976-7 2012 Multivariate analysis revealed a predictive significance for K-Ras mutations with respect to time to progression in patients treated with irinotecan and oxaliplatin as first-line chemotherapy. Irinotecan 138-148 KRAS proto-oncogene, GTPase Homo sapiens 61-66 22909976-11 2012 Also, K-Ras mutation status may predict time to progression in patients treated with irinotecan and oxaliplatin. Irinotecan 85-95 KRAS proto-oncogene, GTPase Homo sapiens 6-11 22922573-0 2012 The synergistic effects of low-dose irinotecan and TRAIL on TRAIL-resistant HT-29 colon carcinoma in vitro and in vivo. Irinotecan 36-46 TNF superfamily member 10 Homo sapiens 60-65 22922573-3 2012 In this study, we investigated the synergistic effects of low-dose irinotecan (CPT-11) and TRAIL on TRAIL-resistant HT-29 colon carcinoma cells and explored potential mechanisms of apoptosis. Irinotecan 67-77 TNF superfamily member 10 Homo sapiens 100-105 22922573-3 2012 In this study, we investigated the synergistic effects of low-dose irinotecan (CPT-11) and TRAIL on TRAIL-resistant HT-29 colon carcinoma cells and explored potential mechanisms of apoptosis. Irinotecan 79-85 TNF superfamily member 10 Homo sapiens 100-105 22922573-8 2012 Our results showed that the antitumor effect of TRAIL could be enhanced significantly by low-dose CPT-11 on TRAIL-resistant HT-29 cells both in vitro and in vivo. Irinotecan 98-104 TNF superfamily member 10 Homo sapiens 48-53 22922573-8 2012 Our results showed that the antitumor effect of TRAIL could be enhanced significantly by low-dose CPT-11 on TRAIL-resistant HT-29 cells both in vitro and in vivo. Irinotecan 98-104 TNF superfamily member 10 Homo sapiens 108-113 22922573-9 2012 The synergistic apoptotic effect of CPT-11 and TRAIL was proposed to be mediated by upregulating DR5 mRNA expression and increasing expression of Bax and caspase-9 proteins. Irinotecan 36-42 TNF receptor superfamily member 10b Homo sapiens 97-100 22922573-9 2012 The synergistic apoptotic effect of CPT-11 and TRAIL was proposed to be mediated by upregulating DR5 mRNA expression and increasing expression of Bax and caspase-9 proteins. Irinotecan 36-42 BCL2 associated X, apoptosis regulator Homo sapiens 146-149 22922573-9 2012 The synergistic apoptotic effect of CPT-11 and TRAIL was proposed to be mediated by upregulating DR5 mRNA expression and increasing expression of Bax and caspase-9 proteins. Irinotecan 36-42 caspase 9 Homo sapiens 154-163 22898888-6 2012 Further analyses showed that CPT-11 induced in both types of cells DNA damage and activated stress response pathways including p53 and p16 but with varying activity of stress kinase p38 and selected stress-associated microRNAs. Irinotecan 29-35 tumor protein p53 Homo sapiens 127-130 23387089-1 2012 In this work, we developed PEO-PPO-PEO micelles loaded with irinotecan hydrochloride (CPT-11) using breast cancer resistance protein (BCRP) inhibitory material PEO20-PPO70-PEO20, and studied its mechanism of decreasing CPT-11 induced delayed diarrhea and intestinal toxicity. Irinotecan 86-92 ATP binding cassette subfamily G member 2 Canis lupus familiaris 134-138 23387089-4 2012 The results showed that the obtained micelles could decrease the biliary excretion of CPT-11, ameliorate delayed diarrhea and intestinal toxicity of rats through inhibiting BCRP-mediated CPT-11 efflux. Irinotecan 86-92 ATP binding cassette subfamily G member 2 Canis lupus familiaris 173-177 23387089-4 2012 The results showed that the obtained micelles could decrease the biliary excretion of CPT-11, ameliorate delayed diarrhea and intestinal toxicity of rats through inhibiting BCRP-mediated CPT-11 efflux. Irinotecan 187-193 ATP binding cassette subfamily G member 2 Canis lupus familiaris 173-177 22929806-10 2012 The antitumor activity of both gemcitabine and irinotecan were significantly enhanced by CCT244747 in several human tumor xenografts, giving concomitant biomarker modulation indicative of CHK1 inhibition. Irinotecan 47-57 checkpoint kinase 1 Homo sapiens 188-192 22898888-6 2012 Further analyses showed that CPT-11 induced in both types of cells DNA damage and activated stress response pathways including p53 and p16 but with varying activity of stress kinase p38 and selected stress-associated microRNAs. Irinotecan 29-35 mitogen-activated protein kinase 14 Homo sapiens 182-185 22898888-6 2012 Further analyses showed that CPT-11 induced in both types of cells DNA damage and activated stress response pathways including p53 and p16 but with varying activity of stress kinase p38 and selected stress-associated microRNAs. Irinotecan 29-35 cyclin dependent kinase inhibitor 2A Homo sapiens 135-138 23335549-0 2012 Electrolyte abnormalities due to irinotecan administration in metastatic HER-2 positive breast cancer patients. Irinotecan 33-43 erb-b2 receptor tyrosine kinase 2 Homo sapiens 73-78 22770988-4 2012 Interestingly, when gef gene expression was combined with drugs of choice in the clinical treatment of colon cancer (5-fluorouracil, oxaliplatin and irinotecan), a strong synergistic effect was observed with approximately a 15-20% enhancement of the antiproliferative effect. Irinotecan 149-159 Rho/Rac guanine nucleotide exchange factor 2 Homo sapiens 20-23 22167582-9 2012 Increased apoptosis monitored by caspase-3 was observed after day 15 with irinotecan treatment. Irinotecan 74-84 caspase 3 Mus musculus 33-42 22614107-5 2012 Using the MTT assay, we found that zafirlukast enhanced the cytotoxicity of several anticancer drugs that are substrates of breast cancer resistance proteins (BCRP/ABCG2), including mitoxantrone and SN-38. Irinotecan 199-204 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 164-169 22797812-13 2012 Transfection of siRNA against WRN increased the sensitivity of the cells to CPT-11. Irinotecan 88-94 WRN RecQ like helicase Homo sapiens 30-33 22797812-14 2012 Aberrant DNA hypermethylation of WRN also increased the sensitivity of cervical cancer cells to CPT-11. Irinotecan 108-114 WRN RecQ like helicase Homo sapiens 45-48 22797812-16 2012 This is the first report to show a relationship between the methylation of the WRN gene and sensitivity to CPT-11 in gynecological cancers. Irinotecan 119-125 WRN RecQ like helicase Homo sapiens 91-94 23202910-8 2012 The HB1.F3.CE, HB.F3.CD, or HB1.F3.CD.IFN-beta cells significantly reduced the LNCaP cell viability in the presence of the prodrugs 5-FC or CPT-11. Irinotecan 140-146 histocompatibility minor HB-1 Homo sapiens 4-7 23202910-8 2012 The HB1.F3.CE, HB.F3.CD, or HB1.F3.CD.IFN-beta cells significantly reduced the LNCaP cell viability in the presence of the prodrugs 5-FC or CPT-11. Irinotecan 140-146 histocompatibility minor HB-1 Homo sapiens 28-31 23202910-8 2012 The HB1.F3.CE, HB.F3.CD, or HB1.F3.CD.IFN-beta cells significantly reduced the LNCaP cell viability in the presence of the prodrugs 5-FC or CPT-11. Irinotecan 140-146 interferon beta 1 Homo sapiens 38-46 22750060-4 2012 VKJP1 and VKJP3 significantly sensitized ABCG2-expressing cells to established substrates of ABCG2 including mitoxantrone, SN-38, and doxorubicin in a concentration-dependent manner, but not to the non-ABCG2 substrate cisplatin. Irinotecan 123-128 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 41-46 22750060-4 2012 VKJP1 and VKJP3 significantly sensitized ABCG2-expressing cells to established substrates of ABCG2 including mitoxantrone, SN-38, and doxorubicin in a concentration-dependent manner, but not to the non-ABCG2 substrate cisplatin. Irinotecan 123-128 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 93-98 22750060-4 2012 VKJP1 and VKJP3 significantly sensitized ABCG2-expressing cells to established substrates of ABCG2 including mitoxantrone, SN-38, and doxorubicin in a concentration-dependent manner, but not to the non-ABCG2 substrate cisplatin. Irinotecan 123-128 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 93-98 23323463-8 2012 The prodrug irinotecan is biotransformed by carboxylesterase into its active metabolite SN-38, which is inactivated by UGT1 into the inactive compound SN-38G. Irinotecan 12-22 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 119-123 23323463-8 2012 The prodrug irinotecan is biotransformed by carboxylesterase into its active metabolite SN-38, which is inactivated by UGT1 into the inactive compound SN-38G. Irinotecan 88-93 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 119-123 23323463-8 2012 The prodrug irinotecan is biotransformed by carboxylesterase into its active metabolite SN-38, which is inactivated by UGT1 into the inactive compound SN-38G. Irinotecan 151-156 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 119-123 22192364-3 2012 Anti-EGFR agents can be added to first-line FOLFIRI (5-fluorouracil, leucovorin [folinic acid], irinotecan) or FOLFOX (5-fluorouracil, leucovorin [folinic acid], oxaliplatin) in patients whose tumors express wild-type KRAS. Irinotecan 96-106 epidermal growth factor receptor Homo sapiens 5-9 23091446-8 2012 RESULTS: Anti-cancer drug-induced cell death was significantly enhanced by knockdown of MKP-1, and this effect was most prominent in cells treated with irinotecan and etoposide. Irinotecan 152-162 dual specificity phosphatase 1 Homo sapiens 88-93 21706149-0 2012 Biweekly cetuximab plus irinotecan as second-line chemotherapy for patients with irinotecan-refractory and KRAS wild-type metastatic colorectal cancer according to epidermal growth factor receptor expression status. Irinotecan 24-34 KRAS proto-oncogene, GTPase Homo sapiens 107-111 22722827-2 2012 We previously found that a gefitinib-resistant cell line, PC-9/Met in which MET (MNNG-HOS transforming gene) is amplified, was more sensitive than its parent cell line (PC-9) to 7-ethyl-10-hydroxy-camptothecin (SN-38), an active metabolite of irinotecan. Irinotecan 211-216 proprotein convertase subtilisin/kexin type 9 Homo sapiens 58-62 22722827-2 2012 We previously found that a gefitinib-resistant cell line, PC-9/Met in which MET (MNNG-HOS transforming gene) is amplified, was more sensitive than its parent cell line (PC-9) to 7-ethyl-10-hydroxy-camptothecin (SN-38), an active metabolite of irinotecan. Irinotecan 243-253 proprotein convertase subtilisin/kexin type 9 Homo sapiens 58-62 22722827-4 2012 METHODS: The sensitivity of PC-9 and PC-9/Met to SN-38 was assessed by performing water soluble tetrazolium salt (WST-1) assays. Irinotecan 49-54 proprotein convertase subtilisin/kexin type 9 Homo sapiens 28-41 22722827-9 2012 CONCLUSIONS: The increased sensitivity of PC-9/Met cells to SN-38 compared with that of PC-9 cells was partially because of topo I activities resulting from increased topo I mRNA and protein expression caused by MET signaling. Irinotecan 60-65 proprotein convertase subtilisin/kexin type 9 Homo sapiens 42-46 22960892-1 2012 BACKGROUND & OBJECTIVES: Genetic polymorphisms of uridine diphosphate glucuronyltransferase 1A1 (UGT1A1) have been associated with a wide variation of responses among patients prescribed with irinotecan. Irinotecan 196-206 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 101-107 22960892-3 2012 The objective of this study was to investigate the prevalence of three different variants of UGT1A1 (UGT1A1*6, UGT1A1*27 and UGT1A1*28), which are associated with reduced enzyme activity and increased irinotecan toxicity, in the three main ethnic groups in Malaysia (Malays, Chinese and Indians). Irinotecan 201-211 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-99 22960892-3 2012 The objective of this study was to investigate the prevalence of three different variants of UGT1A1 (UGT1A1*6, UGT1A1*27 and UGT1A1*28), which are associated with reduced enzyme activity and increased irinotecan toxicity, in the three main ethnic groups in Malaysia (Malays, Chinese and Indians). Irinotecan 201-211 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 101-107 22960892-3 2012 The objective of this study was to investigate the prevalence of three different variants of UGT1A1 (UGT1A1*6, UGT1A1*27 and UGT1A1*28), which are associated with reduced enzyme activity and increased irinotecan toxicity, in the three main ethnic groups in Malaysia (Malays, Chinese and Indians). Irinotecan 201-211 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 101-107 22960892-3 2012 The objective of this study was to investigate the prevalence of three different variants of UGT1A1 (UGT1A1*6, UGT1A1*27 and UGT1A1*28), which are associated with reduced enzyme activity and increased irinotecan toxicity, in the three main ethnic groups in Malaysia (Malays, Chinese and Indians). Irinotecan 201-211 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 101-107 22960892-12 2012 Our results suggest that genotyping of UUGT1A1*6, UGT1A1*28 and UGT1A1*27 need to be performed before patients are prescribed with irinotecan due to their high prevalence of allelic variant which could lead to adverse drug reaction. Irinotecan 131-141 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 40-46 21706149-11 2012 CONCLUSIONS: Biweekly cetuximab plus irinotecan as second-line treatment showed significant anti-tumor activity in patients with irinotecan-refractory mCRC and WT-KRAS regardless of EGFR expression status. Irinotecan 37-47 KRAS proto-oncogene, GTPase Homo sapiens 163-167 21706149-1 2012 BACKGROUND: Cetuximab plus irinotecan has been shown to be effective in metastatic colorectal cancer (mCRC) patients with wild-type (WT) KRAS and positive EGFR expressions (EGFR+). Irinotecan 27-37 KRAS proto-oncogene, GTPase Homo sapiens 137-141 21706149-1 2012 BACKGROUND: Cetuximab plus irinotecan has been shown to be effective in metastatic colorectal cancer (mCRC) patients with wild-type (WT) KRAS and positive EGFR expressions (EGFR+). Irinotecan 27-37 epidermal growth factor receptor Homo sapiens 155-159 21706149-1 2012 BACKGROUND: Cetuximab plus irinotecan has been shown to be effective in metastatic colorectal cancer (mCRC) patients with wild-type (WT) KRAS and positive EGFR expressions (EGFR+). Irinotecan 27-37 epidermal growth factor receptor Homo sapiens 173-177 22535688-2 2012 Intestinal bacteria convert the glucuronidated metabolite back to the toxic SN-38 using beta-glucuronidase (GUS), resulting in debilitating diarrhea. Irinotecan 76-81 glucuronidase beta Homo sapiens 88-106 22614251-13 2012 Neoadjuvant chemotherapy with irinotecan and nedaplatin was an effective and well-tolerated treatment for patients with bulky stage Ib2 to IIb squamous cell carcinoma of the uterine cervix. Irinotecan 30-40 mitogen-activated protein kinase 8 interacting protein 2 Homo sapiens 144-147 22535688-2 2012 Intestinal bacteria convert the glucuronidated metabolite back to the toxic SN-38 using beta-glucuronidase (GUS), resulting in debilitating diarrhea. Irinotecan 76-81 glucuronidase beta Homo sapiens 108-111 22535688-3 2012 Inhibiting GUS activity may relieve this side effect of irinotecan. Irinotecan 56-66 glucuronidase beta Homo sapiens 11-14 22665525-1 2012 The topoisomerase I inhibitor irinotecan is used to treat advanced colorectal cancer and has been shown to have p53-independent anticancer activity. Irinotecan 30-40 tumor protein p53 Homo sapiens 112-115 22665525-2 2012 The aim of this study was to identify the p53-independent signaling mechanisms activated by irinotecan. Irinotecan 92-102 tumor protein p53 Homo sapiens 42-45 22665525-3 2012 Transcriptional profiling of isogenic HCT116 p53 wild-type and p53 null cells was carried out following treatment with the active metabolite of irinotecan, SN38. Irinotecan 144-154 tumor protein p53 Homo sapiens 45-48 22665525-3 2012 Transcriptional profiling of isogenic HCT116 p53 wild-type and p53 null cells was carried out following treatment with the active metabolite of irinotecan, SN38. Irinotecan 144-154 tumor protein p53 Homo sapiens 63-66 22535333-10 2012 CONCLUSION: The impact of increased risk of toxicity attributed to the UGT1A variants may be offset by irinotecan in clinical practice by dose reduction or the use of colony-stimulating factor. Irinotecan 103-113 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 71-76 22112971-0 2012 Analysis of HER-3, insulin growth factor-1, nuclear factor-kB and epidermal growth factor receptor gene copy number in the prediction of clinical outcome for K-RAS wild-type colorectal cancer patients receiving irinotecan-cetuximab. Irinotecan 211-221 epidermal growth factor receptor Homo sapiens 66-98 21898389-4 2012 Use of KRAS testing (assuming KRAS mutant patients receive only irinotecan) was equally effective and saved $12,428 per patient in the United States. Irinotecan 64-74 KRAS proto-oncogene, GTPase Homo sapiens 7-11 22647487-0 2012 MAPK14/p38alpha confers irinotecan resistance to TP53-defective cells by inducing survival autophagy. Irinotecan 24-34 mitogen-activated protein kinase 14 Homo sapiens 0-6 22647487-0 2012 MAPK14/p38alpha confers irinotecan resistance to TP53-defective cells by inducing survival autophagy. Irinotecan 24-34 mitogen-activated protein kinase 14 Homo sapiens 7-15 22647487-0 2012 MAPK14/p38alpha confers irinotecan resistance to TP53-defective cells by inducing survival autophagy. Irinotecan 24-34 tumor protein p53 Homo sapiens 49-53 22233275-3 2012 Val-SN-38 improved intracellular accumulation approximately 5-fold in MCF7 cells, compared with SN-38, and rather than changes in membrane permeability, the amino acid transporter ATB(0,+) played a role, whereas the dipeptide transporter PEPT1 did not. Irinotecan 4-9 solute carrier family 15 member 1 Homo sapiens 238-243 22747970-4 2012 As the third line chemotherapy, a combination therapy of cetuximab with irinotecan was given, which markedly reduced his levels of serum CEA, and the size and number of liver tumors. Irinotecan 72-82 CEA cell adhesion molecule 3 Homo sapiens 137-140 22415148-1 2012 PURPOSE: We conducted a phase I trial of irinotecan (CPT-11), a topoisomerase I inhibitor, combined with amrubicin, a topoisomerase II inhibitor, with recombinant human granulocyte colony-stimulating factor (rhG-CSF) support to overcome the neutropenia associated with this particular combination. Irinotecan 41-51 colony stimulating factor 3 Homo sapiens 169-206 22415148-1 2012 PURPOSE: We conducted a phase I trial of irinotecan (CPT-11), a topoisomerase I inhibitor, combined with amrubicin, a topoisomerase II inhibitor, with recombinant human granulocyte colony-stimulating factor (rhG-CSF) support to overcome the neutropenia associated with this particular combination. Irinotecan 53-59 colony stimulating factor 3 Homo sapiens 169-206 22521243-0 2012 ALDH+ tumor-initiating cells exhibiting gain in NOTCH1 gene copy number have enhanced regrowth sensitivity to a gamma-secretase inhibitor and irinotecan in colorectal cancer. Irinotecan 142-152 notch receptor 1 Homo sapiens 48-54 22764771-5 2012 Important examples include ketoconazole inhibition of hepatic CYP3A4 in order to increase systemic exposure to docetaxel, irinotecan and etoposide, and cyclosporine inhibition of intestinal ATP-binding cassette transporters in order to decrease the toxicity of irinotecan and increase the bioavailability of oral docetaxel, paclitaxel and topotecan. Irinotecan 122-132 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 62-68 22764771-5 2012 Important examples include ketoconazole inhibition of hepatic CYP3A4 in order to increase systemic exposure to docetaxel, irinotecan and etoposide, and cyclosporine inhibition of intestinal ATP-binding cassette transporters in order to decrease the toxicity of irinotecan and increase the bioavailability of oral docetaxel, paclitaxel and topotecan. Irinotecan 261-271 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 62-68 22213127-2 2012 In GS the UGT1A1*28 variant reduces bilirubin conjugation by 70% and is associated with irinotecan and protease inhibitor side effects. Irinotecan 88-98 UDP glucuronosyltransferase 1 family, polypeptide A1 Mus musculus 10-16 25436684-0 2012 Inhibition of Stat3 by peptide aptamer rS3-PA enhances growth suppressive effects of irinotecan on colorectal cancer cells. Irinotecan 85-95 signal transducer and activator of transcription 3 Homo sapiens 14-19 25436684-8 2012 Irinotecan has been used as a chemotherapeutic agent either as a single agent or in combination with 5-fluorouracil and targeted therapies directed against the epidermal growth factor receptor, such as cetuximab. Irinotecan 0-10 epidermal growth factor receptor Homo sapiens 160-192 22521243-10 2012 These results indicate the combination of PF-03084014 and irinotecan may be effective in reducing tumor recurrence in CRC patients whose tumors exhibit elevated levels of the Notch pathway. Irinotecan 58-68 notch receptor 1 Homo sapiens 175-180 22741034-0 2012 Bax expression is predictive of favorable clinical outcome in chemonaive advanced gastric cancer patients treated with capecitabine, oxaliplatin, and irinotecan regimen. Irinotecan 150-160 BCL2 associated X, apoptosis regulator Homo sapiens 0-3 22676194-0 2012 Impact of the UGT1A1*28 allele on response to irinotecan: a systematic review and meta-analysis. Irinotecan 46-56 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 14-20 22676194-1 2012 AIM: Pre-emptive irinotecan dose reduction for UGT1A1*28 homozygotes may result in reduced risk of severe neutropenia and diarrhea. Irinotecan 17-27 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 47-53 22676194-3 2012 Whether UGT1A1*28 genotype is associated with irinotecan response therefore is an important gap in existing knowledge to inform clinical utility. Irinotecan 46-56 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 8-14 22676194-4 2012 MATERIALS & METHODS: A systematic review and meta-analysis was performed to analyze the difference in objective response rate (ORR) between irinotecan-administered cancer patients with different UGT1A1*28 genotypes: *28/*28 (homozygous variant), *1/*28 (heterozygous variant) or *1/*1 (wild-type). Irinotecan 144-154 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 199-205 22676194-5 2012 The effect of irinotecan dose on the association between UGT1A1*28 and ORR was also assessed. Irinotecan 14-24 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 57-63 22426819-3 2012 Breast cancer resistance protein (BCRP)/ABCG2, a drug efflux pump, confers resistance to multiple anticancer agents such as SN-38 and topotecan. Irinotecan 136-141 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 0-32 22426819-3 2012 Breast cancer resistance protein (BCRP)/ABCG2, a drug efflux pump, confers resistance to multiple anticancer agents such as SN-38 and topotecan. Irinotecan 136-141 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 34-38 22426819-3 2012 Breast cancer resistance protein (BCRP)/ABCG2, a drug efflux pump, confers resistance to multiple anticancer agents such as SN-38 and topotecan. Irinotecan 136-141 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 40-45 22426819-6 2012 LY294002, but not wortmannin, reversed the BCRP-mediated SN-38 and topotecan resistance. Irinotecan 69-74 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 43-47 22741034-2 2012 The purpose of this study was to evaluate the prognostic role of Bax in patients with advanced gastric cancer treated with triplet chemotherapy COI regimen (capecitabine, oxaliplatin, and irinotecan). Irinotecan 188-198 BCL2 associated X, apoptosis regulator Homo sapiens 65-68 22471369-7 2012 Increased activity of caspase-3 and caspase-9 was observed after treating with panaxadiol and irinotecan. Irinotecan 94-104 caspase 3 Homo sapiens 22-31 22438565-11 2012 In both genders, AR was the main driver of TUBB3/TUBB6 expression, as constitutive silencing of AR was associated with downregulation of TUBB3/TUBB6 expression and increased sensitivity to oxaliplatin and SN-38. Irinotecan 205-210 androgen receptor Homo sapiens 96-98 22438565-11 2012 In both genders, AR was the main driver of TUBB3/TUBB6 expression, as constitutive silencing of AR was associated with downregulation of TUBB3/TUBB6 expression and increased sensitivity to oxaliplatin and SN-38. Irinotecan 205-210 tubulin beta 3 class III Homo sapiens 137-142 22328001-10 2012 Knockdown of CIP2A decreased the resistance of the cells to 5-fluorouracil, oxaliplatin, and SN38 (an active metabolite of irinotecan). Irinotecan 123-133 cellular inhibitor of PP2A Homo sapiens 13-18 22069124-9 2012 Thus, targeted therapies directed against calpain 2 may represent a novel strategy to enhance the anti-cancer efficacy of CPT-11. Irinotecan 122-128 calpain 2 Homo sapiens 42-51 22516947-0 2012 Phase II and UGT1A1 genotype study of irinotecan dose escalation as salvage therapy for advanced gastric cancer. Irinotecan 38-48 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 13-19 22069124-0 2012 Calpain 2-dependent IkappaBalpha degradation mediates CPT-11 secondary resistance in colorectal cancer xenografts. Irinotecan 54-60 calpain 2 Homo sapiens 0-9 22069124-0 2012 Calpain 2-dependent IkappaBalpha degradation mediates CPT-11 secondary resistance in colorectal cancer xenografts. Irinotecan 54-60 NFKB inhibitor alpha Homo sapiens 20-32 22069124-6 2012 The decrease of NF-kappaB activation in HT-29R and RSN cells by stable transfections with the super-repressor form of IkappaBalpha augmented their sensitivity to CPT-11. Irinotecan 162-168 NFKB inhibitor alpha Homo sapiens 118-130 22069124-8 2012 SiRNA silencing of calpain 2 but not of S100A10 and/or annexin A2, resulted in a decrease in NF-kappaB activation, an increase in cellular levels of IkappaBalpha and a partial restoration of the CPT-11 sensitivity in both HT-29R and RSN cells, suggesting that calpain 2-dependent IkappaBalpha degradation mediates CPT-11 secondary resistance. Irinotecan 195-201 calpain 2 Homo sapiens 19-28 22069124-8 2012 SiRNA silencing of calpain 2 but not of S100A10 and/or annexin A2, resulted in a decrease in NF-kappaB activation, an increase in cellular levels of IkappaBalpha and a partial restoration of the CPT-11 sensitivity in both HT-29R and RSN cells, suggesting that calpain 2-dependent IkappaBalpha degradation mediates CPT-11 secondary resistance. Irinotecan 314-320 calpain 2 Homo sapiens 19-28 22471369-7 2012 Increased activity of caspase-3 and caspase-9 was observed after treating with panaxadiol and irinotecan. Irinotecan 94-104 caspase 9 Homo sapiens 36-45 22471369-8 2012 The synergistic apoptotic effects were supported by docking analysis, which demonstrated that panaxadiol and irinotecan bound two different chains of the caspase-3 protein. Irinotecan 109-119 caspase 3 Homo sapiens 154-163 22471369-9 2012 CONCLUSIONS: Data from this study suggested that caspase-3- and caspase-9-mediated apoptosis may play an important role in the panaxadiol enhanced antiproliferative effects of irinotecan on human colorectal cancer cells. Irinotecan 176-186 caspase 3 Homo sapiens 49-73 22490361-3 2012 METHODS: We tested the interaction between phosphorylated AKT and MAPK expression in colorectal tumours and corresponding metastases and global outcome in K-RAS wild type patients receiving irinotecan-cetuximab. Irinotecan 190-200 KRAS proto-oncogene, GTPase Homo sapiens 155-160 22264223-8 2012 In third-line studies cetuximab-irinotecan is associated with a significant advantage in ORR, mTTP and OS. Irinotecan 32-42 zinc finger protein 36 Mus musculus 94-98 22264223-10 2012 In third-line studies cetuximab-irinotecan is associated with a significant advantage in ORR, mTTP and OS. Irinotecan 32-42 zinc finger protein 36 Mus musculus 94-98 22237959-0 2012 Ugt1a is required for the protective effect of selenium against irinotecan-induced toxicity. Irinotecan 64-74 Ugt1a@ Rattus norvegicus 0-5 22101361-4 2012 The aim of this study was to investigate the role of NO on the pathogenesis of intestinal mucositis, as well as the participation of cytokines upon inducible nitric oxide synthase (iNOS) expression in irinotecan-induced intestinal mucositis. Irinotecan 201-211 nitric oxide synthase 2, inducible Mus musculus 148-179 22101361-4 2012 The aim of this study was to investigate the role of NO on the pathogenesis of intestinal mucositis, as well as the participation of cytokines upon inducible nitric oxide synthase (iNOS) expression in irinotecan-induced intestinal mucositis. Irinotecan 201-211 nitric oxide synthase 2, inducible Mus musculus 181-185 22101361-9 2012 Additionally, increased MPO and iNOS activity and iNOS immunoexpression were found in WT animals treated with irinotecan. Irinotecan 110-120 myeloperoxidase Mus musculus 24-27 22237959-4 2012 The objective of this study is to determine the role of a specific group of uridine diphosphate glucuronosyltransferases, which is coded by UGT1A, in detoxification process of irinotecan as well as selenium-associated protective effect against irinotecan-induced toxicity. Irinotecan 176-186 Ugt1a@ Rattus norvegicus 140-145 22101361-9 2012 Additionally, increased MPO and iNOS activity and iNOS immunoexpression were found in WT animals treated with irinotecan. Irinotecan 110-120 nitric oxide synthase 2, inducible Mus musculus 32-36 22101361-9 2012 Additionally, increased MPO and iNOS activity and iNOS immunoexpression were found in WT animals treated with irinotecan. Irinotecan 110-120 nitric oxide synthase 2, inducible Mus musculus 50-54 22237959-11 2012 CONCLUSION: These data support the hypothesis that expression of UGT1A is critical for 5-methylselenocysteine to exert its protective effect against irinotecan-induced toxicity. Irinotecan 149-159 Ugt1a@ Rattus norvegicus 65-70 22237959-4 2012 The objective of this study is to determine the role of a specific group of uridine diphosphate glucuronosyltransferases, which is coded by UGT1A, in detoxification process of irinotecan as well as selenium-associated protective effect against irinotecan-induced toxicity. Irinotecan 244-254 Ugt1a@ Rattus norvegicus 140-145 22101361-11 2012 Moreover, through western blot, we verified that infliximab and pentoxifylline significantly inhibited irinotecan-induced iNOS expression. Irinotecan 103-113 nitric oxide synthase 2, inducible Mus musculus 122-126 22101361-12 2012 In addition, cytokine concentration was found only partially decreased in irinotecan-treated iNOS(-/-) mice when compared with wild-type animals that were given irinotecan. Irinotecan 74-84 nitric oxide synthase 2, inducible Mus musculus 93-97 22237959-9 2012 The enhanced sensitivity was specific to irinotecan and was not observed with other chemotherapeutic agents, such as docetaxel and cisplatin, where Ugt1a is not required for their metabolism. Irinotecan 41-51 Ugt1a@ Rattus norvegicus 148-153 22202118-0 2012 Human organic cation transporter 1 is expressed in lymphoma cells and increases susceptibility to irinotecan and paclitaxel. Irinotecan 98-108 solute carrier family 22 member 1 Homo sapiens 6-34 21174225-0 2012 Phase II study of combination chemotherapy with biweekly cetuximab and irinotecan for wild-type KRAS metastatic colorectal cancer refractory to irinotecan, oxaliplatin, and fluoropyrimidines. Irinotecan 71-81 KRAS proto-oncogene, GTPase Homo sapiens 96-100 21174225-12 2012 Combination chemotherapy with biweekly cetuximab and irinotecan was effective for pretreated metastatic colorectal cancer with wild-type KRAS. Irinotecan 53-63 KRAS proto-oncogene, GTPase Homo sapiens 137-141 22446188-5 2012 We therefore tested whether inhibition of Chk1 could potentiate the cytotoxicity of the DNA damaging agent irinotecan in TNBC using xenotransplant tumor models. Irinotecan 107-117 checkpoint kinase 1 Homo sapiens 42-46 22202118-4 2012 Functionally, the antineoplastic agents irinotecan, mitoxantrone, and paclitaxel inhibited the uptake of the organic cation [(3)H]1-methyl-4-pyridinium iodide into OCT1-transfected Chinese hamster ovary model cells, with K(i) values of 1.7, 85, and 50 muM, respectively. Irinotecan 40-50 solute carrier family 22 member 1 Homo sapiens 164-168 22202118-5 2012 Correspondingly, OCT1-positive cell lines and transfectants exhibited significantly higher susceptibilities to the cytotoxic effects of irinotecan and paclitaxel compared with those of OCT1-negative controls. Irinotecan 136-146 solute carrier family 22 member 1 Homo sapiens 17-21 22200670-0 2012 Differential effects of calcium antagonists on ABCG2/BCRP-mediated drug resistance and transport in SN-38-resistant HeLa cells. Irinotecan 112-117 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 47-52 22389470-4 2012 We found that human epidermal growth factor receptor 2 (HER2) expressions are also upregulated in chemoresistant SBC-3/ETP, SBC-3/SN-38, and SBC-3/CDDP cells, compared with chemosensitive SBC-3 cells. Irinotecan 130-135 erb-b2 receptor tyrosine kinase 2 Homo sapiens 20-54 22389470-4 2012 We found that human epidermal growth factor receptor 2 (HER2) expressions are also upregulated in chemoresistant SBC-3/ETP, SBC-3/SN-38, and SBC-3/CDDP cells, compared with chemosensitive SBC-3 cells. Irinotecan 130-135 erb-b2 receptor tyrosine kinase 2 Homo sapiens 56-60 22389470-5 2012 Lapatinib, a tyrosine kinase inhibitor of HER2, could not suppress proliferation of these HER2-positive SCLC cells alone but successfully restored chemosensitivity to etoposide and SN-38 with a clinically applicable concentration. Irinotecan 181-186 erb-b2 receptor tyrosine kinase 2 Homo sapiens 42-46 21176921-10 2012 Claudin-1 protein expression of both the small and large intestine decreased (P < 0.05), occludin protein expression of the small intestine decreased (P < 0.05), and occludin protein expression of the large intestine had decreasing tendency (P = 0.07) in irinotecan treated rats. Irinotecan 261-271 occludin Rattus norvegicus 172-180 21176921-11 2012 In irinotecan treated rats, claudin-1 mRNA of the small intestine decreased (P < 0.05), claudin-1 mRNA of large intestine had a tendency to decrease (P = 0.05), occludin mRNA of both small and large intestine decreased (P < 0.05). Irinotecan 3-13 claudin 1 Rattus norvegicus 28-37 21176921-11 2012 In irinotecan treated rats, claudin-1 mRNA of the small intestine decreased (P < 0.05), claudin-1 mRNA of large intestine had a tendency to decrease (P = 0.05), occludin mRNA of both small and large intestine decreased (P < 0.05). Irinotecan 3-13 claudin 1 Rattus norvegicus 91-100 21176921-11 2012 In irinotecan treated rats, claudin-1 mRNA of the small intestine decreased (P < 0.05), claudin-1 mRNA of large intestine had a tendency to decrease (P = 0.05), occludin mRNA of both small and large intestine decreased (P < 0.05). Irinotecan 3-13 occludin Rattus norvegicus 164-172 21176921-12 2012 CONCLUSIONS: Irinotecan injures claudin-1 and occludin. Irinotecan 13-23 claudin 1 Rattus norvegicus 32-41 21176921-12 2012 CONCLUSIONS: Irinotecan injures claudin-1 and occludin. Irinotecan 13-23 occludin Rattus norvegicus 46-54 22469241-7 2012 For irinotecan-, oxaliplatin-, and bevacizumab-treated tumors, a significant correlation was established between the radiotracer uptake and caspase-3 immunostaining (r = .8, p < .05; r = .9, p < .001; r = .9, p < .001, respectively). Irinotecan 4-14 caspase 3 Mus musculus 140-149 22399605-0 2012 SN-38 overcomes chemoresistance of colorectal cancer cells induced by hypoxia, through HIF1alpha. Irinotecan 0-5 hypoxia inducible factor 1 subunit alpha Homo sapiens 87-96 22399605-7 2012 This effect was dependent on the significant inhibition of the accumulation of HIF-1alpha in cancer cells cultured under hypoxia by SN-38. Irinotecan 132-137 hypoxia inducible factor 1 subunit alpha Homo sapiens 79-89 22399605-9 2012 CONCLUSION: SN-38, through inhibition of HIF-1alpha can overcome chemoresistance under hypoxic conditions of colon cancer cells. Irinotecan 12-17 hypoxia inducible factor 1 subunit alpha Homo sapiens 41-51 22200670-0 2012 Differential effects of calcium antagonists on ABCG2/BCRP-mediated drug resistance and transport in SN-38-resistant HeLa cells. Irinotecan 112-117 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 53-57 22033674-5 2012 Unexpectedly, the in vivo antitumor activity of irinotecan was closely linked to a downregulation of hypoxia-inducible factor-1alpha (HIF1A) target genes along with an inhibition of HIF1A protein accumulation. Irinotecan 48-58 hypoxia inducible factor 1 subunit alpha Homo sapiens 101-132 22740978-0 2012 Correlation between plasma concentration ratios of SN-38 glucuronide and SN-38 and neutropenia induction in patients with colorectal cancer and wild-type UGT1A1 gene. Irinotecan 51-56 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 154-160 22740978-3 2012 A total of 17 patients with colorectal cancer and wild-type UDP-glucuronosyltransferase (UGT)1A1 gene were enrolled and treated with CPT-11 as part of the FOLFIRI regimen [CPT-11 and fluorouracil (5-FU)]. Irinotecan 133-139 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 89-96 22740978-6 2012 The median, maximum and minimum values of plasma SN-38G/SN-38 ratios were 4.25, 7.09 and 1.03, respectively, indicating that UGT activities are variable among patients with the wild-type UGT1A1 gene. Irinotecan 49-54 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 125-128 22740978-7 2012 The plasma SN-38G/SN-38 ratios decreased with an increase in the trial numbers of chemotherapy (r=0.741, p=0.000669), suggesting that CPT-11 treatment suppresses UGT activity, and the low plasma SN-38G/SN-38 ratios resulted in the induction of greater neutropenia. Irinotecan 11-16 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 162-165 22740978-7 2012 The plasma SN-38G/SN-38 ratios decreased with an increase in the trial numbers of chemotherapy (r=0.741, p=0.000669), suggesting that CPT-11 treatment suppresses UGT activity, and the low plasma SN-38G/SN-38 ratios resulted in the induction of greater neutropenia. Irinotecan 134-140 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 162-165 22740978-9 2012 In conclusion, UGT activity involved in SN-38 metabolism is variable among patients with the wild-type UGT1A1 gene, and each CPT-11 treatment suppresses UGT activity. Irinotecan 40-45 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 15-18 22740978-9 2012 In conclusion, UGT activity involved in SN-38 metabolism is variable among patients with the wild-type UGT1A1 gene, and each CPT-11 treatment suppresses UGT activity. Irinotecan 40-45 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 103-109 22740978-9 2012 In conclusion, UGT activity involved in SN-38 metabolism is variable among patients with the wild-type UGT1A1 gene, and each CPT-11 treatment suppresses UGT activity. Irinotecan 40-45 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 103-106 22740978-9 2012 In conclusion, UGT activity involved in SN-38 metabolism is variable among patients with the wild-type UGT1A1 gene, and each CPT-11 treatment suppresses UGT activity. Irinotecan 125-131 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 15-18 22740978-9 2012 In conclusion, UGT activity involved in SN-38 metabolism is variable among patients with the wild-type UGT1A1 gene, and each CPT-11 treatment suppresses UGT activity. Irinotecan 125-131 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 103-109 22740978-9 2012 In conclusion, UGT activity involved in SN-38 metabolism is variable among patients with the wild-type UGT1A1 gene, and each CPT-11 treatment suppresses UGT activity. Irinotecan 125-131 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 103-106 22033674-5 2012 Unexpectedly, the in vivo antitumor activity of irinotecan was closely linked to a downregulation of hypoxia-inducible factor-1alpha (HIF1A) target genes along with an inhibition of HIF1A protein accumulation. Irinotecan 48-58 hypoxia inducible factor 1 subunit alpha Homo sapiens 134-139 22033674-5 2012 Unexpectedly, the in vivo antitumor activity of irinotecan was closely linked to a downregulation of hypoxia-inducible factor-1alpha (HIF1A) target genes along with an inhibition of HIF1A protein accumulation. Irinotecan 48-58 hypoxia inducible factor 1 subunit alpha Homo sapiens 182-187 22788763-7 2012 Since irinotecan is increasingly being combined with radiotherapy, the potential for these proteins to act as predictive biomarkers for both Top I inhibitors and radiation is proposed, and the possibility of synergistic potentiation of chemoradiation regimes by Tdp1 and/or PARP inhibitors is considered. Irinotecan 6-16 DNA topoisomerase I Homo sapiens 141-146 22147942-11 2012 Additional studies are necessary to assess whether there is any predictive role of BRAF mutation for irinotecan-based therapy. Irinotecan 101-111 B-Raf proto-oncogene, serine/threonine kinase Homo sapiens 83-87 22068817-3 2012 Three chemotherapeutic agents (5-fluorouracil, oxaliplatin and irinotecan) were tested by ATP-CRA and more sensitive agents were selected. Irinotecan 63-73 myotubularin related protein 11 Homo sapiens 94-97 23631285-3 2012 The majority of the toxic manifestation is caused by the insufficient deactivation (glucuronidation) of irinotecan active metabolite SN-38 by UGT1A enzyme. Irinotecan 104-114 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 142-147 23631285-3 2012 The majority of the toxic manifestation is caused by the insufficient deactivation (glucuronidation) of irinotecan active metabolite SN-38 by UGT1A enzyme. Irinotecan 133-138 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 142-147 23631285-6 2012 The mutations in other enzyme systems also play role in the etiopathogenesis of the irinotecan toxicity: CYP3A (cytochrome P-450), ABC family of transmembrane transporters (adenosine-triphosphate binding cassette). Irinotecan 84-94 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 105-110 22213294-1 2012 AIM: We investigated the effects of trastuzumab, an anti-HER2 humanized monoclonal antibody, on DNA breaks induced by SN-38, a topoisomerase-1 inhibitor, in gastric cancer cell lines positive or negative for HER2 expression. Irinotecan 118-123 erb-b2 receptor tyrosine kinase 2 Homo sapiens 57-61 23481572-8 2012 While 5-florouracil did not inhibit the VEGF secretion of HT-29 cell line, irinotecan, oxaliplatin, docetaxel and paclitaxel significantly decreased the levels of secreted VEGF. Irinotecan 75-85 vascular endothelial growth factor A Homo sapiens 172-176 22229270-0 2012 Low-dosage metronomic chemotherapy and angiogenesis: topoisomerase inhibitors irinotecan and mitoxantrone stimulate VEGF-A-mediated angiogenesis. Irinotecan 78-88 vascular endothelial growth factor A Rattus norvegicus 116-122 22229270-4 2012 The aim of the present study was to assess the systemic effect of low-dosage metronomic treatment with either irinotecan or mitoxantrone on angiogenesis induced by VEGF-A. Irinotecan 110-120 vascular endothelial growth factor A Rattus norvegicus 164-170 21896304-6 2012 Cathepsin S expression levels are upregulated in HCT116, LoVo, Colo205 cell lines and HUVECs after exposure to CPT-11 in vitro. Irinotecan 111-117 cathepsin S Homo sapiens 0-11 22379477-4 2012 As hypoglossal nerve activity has been reported to be especially susceptible to cholinergic stimulation and irinotecan can cause cholinergic side effects by binding to and inactivating acetylcholinesterase, we suspect this mechanism to be responsible for irinotecan-induced dysarthria. Irinotecan 108-118 acetylcholinesterase (Cartwright blood group) Homo sapiens 185-205 22379477-4 2012 As hypoglossal nerve activity has been reported to be especially susceptible to cholinergic stimulation and irinotecan can cause cholinergic side effects by binding to and inactivating acetylcholinesterase, we suspect this mechanism to be responsible for irinotecan-induced dysarthria. Irinotecan 255-265 acetylcholinesterase (Cartwright blood group) Homo sapiens 185-205 21040359-6 2012 Hypermethylation of the DYPD gene (encodes the enzyme dihydropyrimidine dehydrogenase) and variation of the uridine diphosphate-glucuronosyltransferase 1A (UGT1A1) gene have predictive value for side effects and the efficacy of 5-fluoruracil and irinotecan, respectively. Irinotecan 246-256 UDP glucuronosyltransferase family 1 member A1 Rattus norvegicus 156-162 23176042-10 2012 Increased topoisomerase I expression and increased EGFR gene copy number as possible predictors of response to irinotecan- and cetuximab-based chemoradiation, respectively, deserve further studies. Irinotecan 111-121 epidermal growth factor receptor Homo sapiens 51-55 21901246-12 2012 In conclusion, silencing of DEXI leads to resistance, but restored expression enhances susceptibility to CPT in vitro and DEXI methylation results in poor response and outcome to CPT-11-based chemotherapy, suggesting that DEXI is a potent therapeutic target and an epigenetic biomarker for the selection of patients more likely to benefit from CPT-11-based chemotherapy. Irinotecan 203-209 Dexi homolog Homo sapiens 40-44 22201120-2 2012 All patients harbored UDP-glucuronosyltransferase (UGT) 1A1*1/*1, *1/*6, or *1/*28 genotypes, which are associated with similar irinotecan pharmacokinetics and responses to FOLFIRI. Irinotecan 128-138 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 22-59 22447115-4 2012 When cells were treated with a typical CYP3A substrate (triazolam), CYP2C9 substrate (diclofenac) or UGT1A1 substrate (SN-38), large amounts of their metabolites were detected in the medium of cells cultured on micro-space cell culture plates. Irinotecan 119-124 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 101-107 22447115-5 2012 The formation of metabolites from triazolam, diclofenac and SN-38 was strongly inhibited by co-treatment with a CYP3A inhibitor (ketoconazole), CYP2C9 inhibitor (sulfaphenazole) and UGT1A1 inhibitor (ketoconazole), respectively. Irinotecan 60-65 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 144-150 22447115-5 2012 The formation of metabolites from triazolam, diclofenac and SN-38 was strongly inhibited by co-treatment with a CYP3A inhibitor (ketoconazole), CYP2C9 inhibitor (sulfaphenazole) and UGT1A1 inhibitor (ketoconazole), respectively. Irinotecan 60-65 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 182-188 21901246-12 2012 In conclusion, silencing of DEXI leads to resistance, but restored expression enhances susceptibility to CPT in vitro and DEXI methylation results in poor response and outcome to CPT-11-based chemotherapy, suggesting that DEXI is a potent therapeutic target and an epigenetic biomarker for the selection of patients more likely to benefit from CPT-11-based chemotherapy. Irinotecan 203-209 Dexi homolog Homo sapiens 146-150 21901246-12 2012 In conclusion, silencing of DEXI leads to resistance, but restored expression enhances susceptibility to CPT in vitro and DEXI methylation results in poor response and outcome to CPT-11-based chemotherapy, suggesting that DEXI is a potent therapeutic target and an epigenetic biomarker for the selection of patients more likely to benefit from CPT-11-based chemotherapy. Irinotecan 203-209 Dexi homolog Homo sapiens 146-150 22946345-2 2012 METHODS: Patients with RSCLC were treated with nedaplatin (NP) at 50 mg/m2 and irinotecan (CPT) at 50 mg/m2 on days 1 and 8 every 4 weeks for four cycles. Irinotecan 79-89 choline phosphotransferase 1 Homo sapiens 91-94 22039078-4 2012 When SN-38 was stably linked to the anti-CD22 conjugate, its potency was reduced 40- to 55-fold. Irinotecan 5-10 CD22 molecule Homo sapiens 41-45 22862168-0 2012 Enhanced chemosensitivity to CPT-11 in colorectal carcinoma xenografts by small hairpin RNA interference targeting PLK1. Irinotecan 29-35 polo like kinase 1 Homo sapiens 116-120 22862168-7 2012 In vivo, employment of shRNA targeting PLK1 improved the sensitivity to treat SW620 nude mouse model toward CPT-11. Irinotecan 108-114 polo like kinase 1 Mus musculus 39-43 22862168-10 2012 Taken together, combination of PLK1-specific shRNA interference with low-dose CPT-11 triggered a antitumor efficacy and represented a potential strategy to treat colon cancer. Irinotecan 78-84 polo like kinase 1 Homo sapiens 31-35 22508373-0 2012 A phase I study of infusional 5-fluorouracil, leucovorin, oxaliplatin and irinotecan in Japanese patients with advanced colorectal cancer who harbor UGT1A1*1/*1,*1/*6 or *1/*28. Irinotecan 74-84 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 149-155 22740861-0 2012 Sequential irinotecan hydrochloride/S-1 for S-1-resistant inoperable gastric cancer: A feasibility study. Irinotecan 11-21 proteasome 26S subunit, non-ATPase 1 Homo sapiens 44-47 22740861-1 2012 Irinotecan hydrochloride (CPT-11) is reported to be involved in the downregulation of thymidylate synthase (TS), a target molecule of 5-fluorouracil (5-FU) and oral fluoropyrimidine S-1. Irinotecan 0-24 thymidylate synthetase Homo sapiens 86-106 22740861-1 2012 Irinotecan hydrochloride (CPT-11) is reported to be involved in the downregulation of thymidylate synthase (TS), a target molecule of 5-fluorouracil (5-FU) and oral fluoropyrimidine S-1. Irinotecan 0-24 proteasome 26S subunit, non-ATPase 1 Homo sapiens 182-185 22740861-1 2012 Irinotecan hydrochloride (CPT-11) is reported to be involved in the downregulation of thymidylate synthase (TS), a target molecule of 5-fluorouracil (5-FU) and oral fluoropyrimidine S-1. Irinotecan 26-32 thymidylate synthetase Homo sapiens 86-106 22740861-1 2012 Irinotecan hydrochloride (CPT-11) is reported to be involved in the downregulation of thymidylate synthase (TS), a target molecule of 5-fluorouracil (5-FU) and oral fluoropyrimidine S-1. Irinotecan 26-32 proteasome 26S subunit, non-ATPase 1 Homo sapiens 182-185 22740861-2 2012 Therefore, we hypothesized that a preceding administration of CPT-11 against S-1-resistant tumors may recover sensitivity to S-1. Irinotecan 62-68 proteasome 26S subunit, non-ATPase 1 Homo sapiens 77-80 22740861-2 2012 Therefore, we hypothesized that a preceding administration of CPT-11 against S-1-resistant tumors may recover sensitivity to S-1. Irinotecan 62-68 proteasome 26S subunit, non-ATPase 1 Homo sapiens 125-128 22777333-1 2012 OBJECTIVE: S-1 is effective in sequential combination with irinotecan (IRIS) in treating metastatic colorectal cancer. Irinotecan 59-69 proteasome 26S subunit, non-ATPase 1 Homo sapiens 11-14 22206574-7 2011 The loss of cytoplasmic NF-kappa-B in response to irinotecan was diminished by sorafenib, providing evidence for a possible benefit for this drug combination. Irinotecan 50-60 nuclear factor kappa B subunit 1 Homo sapiens 24-34 22927913-10 2012 Viability of both XB130-silenced SGC7901 cells and wild-type cells was suppressed by 5-fluorouracil (5-FU), cisplatin, and irinotecan in a dose-dependent way, but cisplatin and irinotecan were more sensitive against sXB130-silenced GC cells and 5-FU showed higher sensitivity to wild-type cells. Irinotecan 123-133 actin filament associated protein 1 like 2 Homo sapiens 18-23 22927913-10 2012 Viability of both XB130-silenced SGC7901 cells and wild-type cells was suppressed by 5-fluorouracil (5-FU), cisplatin, and irinotecan in a dose-dependent way, but cisplatin and irinotecan were more sensitive against sXB130-silenced GC cells and 5-FU showed higher sensitivity to wild-type cells. Irinotecan 177-187 actin filament associated protein 1 like 2 Homo sapiens 18-23 22293705-0 2012 [Examination of factors affecting adverse reactions and dosage reduction in UGT1A1 genotyped patients: a retrospective survey of irinotecan]. Irinotecan 129-139 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 76-82 22293705-1 2012 Our aim was to clarify the side effects of irinotecan which occurred in patients admitted to Showa University Hospital to investigate whether the UGT1A1 genetic polymorphism status was reflected in the discontinuation or dose reduction of irinotecan. Irinotecan 239-249 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 146-152 22293705-8 2012 Our investigation confirmed that the UGT1A1 genetic polymorphism status of the patients was reflected in the discontinuance or dose reduction of irinotecan. Irinotecan 145-155 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 37-43 22114188-7 2011 IF7 conjugated to the potent anticancer drug SN-38 and injected intravenously into nude mice carrying human colon HCT116 tumors efficiently suppressed tumor growth at low dosages with no apparent side effects. Irinotecan 45-50 olfactory receptor family 2 subfamily V member 1 Mus musculus 0-3 22111927-4 2011 A potent and highly selective isoquinoline CHK1 inhibitor (SAR-020106) was identified, which potentiated the efficacies of irinotecan and gemcitabine in SW620 human colon carcinoma xenografts in nude mice. Irinotecan 123-133 checkpoint kinase 1 Homo sapiens 43-47 21985641-5 2011 UGT1A1 activity correlation analyses using flavonoids-4"-O-glucuronidation vs beta-estradiol-3-glucuronidation (a well-recognized marker for UGT1A1) or vs SN-38 glucuronidation were performed using a bank of HLMs (n = 12) including three UGT1A1-genotyped HLMs (i.e., UGT1A1*1*1, UGT1A1*1*28, and UGT1A1*28*28). Irinotecan 155-160 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 21985641-8 2011 In conclusion, UGT1A1-mediated 4"-O-glucuronidation of 3,3",4"-THF and 3,6,4"-THF was highly correlated with the glucuronidation of estradiol (3-OH) and SN-38. Irinotecan 153-158 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 15-21 21840303-0 2011 Hepatocyte growth factor suppresses the anticancer effect of irinotecan by decreasing the level of active metabolite in HepG2 cells. Irinotecan 61-71 hepatocyte growth factor Homo sapiens 0-24 21840303-1 2011 In the liver, carboxylesterase (CES) converts irinotecan (CPT-11) to its active metabolite SN-38, which exerts anticancer effects. Irinotecan 46-56 carboxylesterase 2 Homo sapiens 14-30 21840303-1 2011 In the liver, carboxylesterase (CES) converts irinotecan (CPT-11) to its active metabolite SN-38, which exerts anticancer effects. Irinotecan 58-64 carboxylesterase 2 Homo sapiens 14-30 21840303-1 2011 In the liver, carboxylesterase (CES) converts irinotecan (CPT-11) to its active metabolite SN-38, which exerts anticancer effects. Irinotecan 91-96 carboxylesterase 2 Homo sapiens 14-30 21840303-2 2011 SN-38 is metabolized to an inactive metabolite SN-38 glucuronide by uridine 5"-diphospho-glucuronosyltransferase 1A1 (UGT1A1). Irinotecan 0-5 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 68-116 21840303-2 2011 SN-38 is metabolized to an inactive metabolite SN-38 glucuronide by uridine 5"-diphospho-glucuronosyltransferase 1A1 (UGT1A1). Irinotecan 0-5 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 118-124 21840303-3 2011 Therefore, single nucleotide polymorphisms (SNPs) of the UGT1A1 gene are responsible for the severe adverse effects associated with the disruption of SN-38 metabolism. Irinotecan 150-155 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 57-63 21840303-4 2011 However, despite having SNPs of the UGT1A1 gene, many patients metabolize SN-38 sufficiently to avoid severe adverse effects. Irinotecan 74-79 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 36-42 21840303-6 2011 The aim of this study was to evaluate whether HGF alters the metabolism of CPT-11, resulting in a reduction in the anticancer effect of CPT11. Irinotecan 75-81 hepatocyte growth factor Homo sapiens 46-49 21840303-7 2011 The cytotoxicity of CPT-11 and SN-38 was evaluated in HepG2 cells pretreated with HGF. Irinotecan 20-26 hepatocyte growth factor Homo sapiens 82-85 21840303-7 2011 The cytotoxicity of CPT-11 and SN-38 was evaluated in HepG2 cells pretreated with HGF. Irinotecan 31-36 hepatocyte growth factor Homo sapiens 82-85 21840303-9 2011 HGF suppressed the cytotoxicity of CPT-11 by decreasing intracellular SN-38 levels that resulted from a decrease in CES2 and an increase in UGT1A1. Irinotecan 35-41 hepatocyte growth factor Homo sapiens 0-3 21840303-9 2011 HGF suppressed the cytotoxicity of CPT-11 by decreasing intracellular SN-38 levels that resulted from a decrease in CES2 and an increase in UGT1A1. Irinotecan 35-41 carboxylesterase 2 Homo sapiens 116-120 21840303-9 2011 HGF suppressed the cytotoxicity of CPT-11 by decreasing intracellular SN-38 levels that resulted from a decrease in CES2 and an increase in UGT1A1. Irinotecan 35-41 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 140-146 21840303-9 2011 HGF suppressed the cytotoxicity of CPT-11 by decreasing intracellular SN-38 levels that resulted from a decrease in CES2 and an increase in UGT1A1. Irinotecan 70-75 hepatocyte growth factor Homo sapiens 0-3 21840303-9 2011 HGF suppressed the cytotoxicity of CPT-11 by decreasing intracellular SN-38 levels that resulted from a decrease in CES2 and an increase in UGT1A1. Irinotecan 70-75 carboxylesterase 2 Homo sapiens 116-120 21840303-9 2011 HGF suppressed the cytotoxicity of CPT-11 by decreasing intracellular SN-38 levels that resulted from a decrease in CES2 and an increase in UGT1A1. Irinotecan 70-75 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 140-146 21840303-12 2011 These results suggest that HGF is a possible causative agent of acquired clinical resistance in chemotherapy with CPT-11 and could be useful as a predictor of clinical resistance. Irinotecan 114-120 hepatocyte growth factor Homo sapiens 27-30 21787264-5 2011 In this view, the uridine diphosphate glucuronosil transferase isoform 1A1 (UGT1A1) was associated with significant changes in disposition of CPT-11 and its metabolites, and consequently with treatment-induced toxicities. Irinotecan 142-148 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 18-74 21787264-5 2011 In this view, the uridine diphosphate glucuronosil transferase isoform 1A1 (UGT1A1) was associated with significant changes in disposition of CPT-11 and its metabolites, and consequently with treatment-induced toxicities. Irinotecan 142-148 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 76-82 21787264-6 2011 However, the complex pharmacokinetics of irinotecan and the involvement of several enzymes other than UGT (i.e., carboxyl estherases, CYP450 isoforms), and transmembrane transporters (ABCB1, ABCC1, ABCG2, SLCO1B1) make difficult the identification of patients with an optimal sensitivity and specificity, and a large part of variability among patients still remains unexplained. Irinotecan 41-51 ATP binding cassette subfamily B member 1 Homo sapiens 184-189 21787264-6 2011 However, the complex pharmacokinetics of irinotecan and the involvement of several enzymes other than UGT (i.e., carboxyl estherases, CYP450 isoforms), and transmembrane transporters (ABCB1, ABCC1, ABCG2, SLCO1B1) make difficult the identification of patients with an optimal sensitivity and specificity, and a large part of variability among patients still remains unexplained. Irinotecan 41-51 ATP binding cassette subfamily C member 1 Homo sapiens 191-196 21787264-6 2011 However, the complex pharmacokinetics of irinotecan and the involvement of several enzymes other than UGT (i.e., carboxyl estherases, CYP450 isoforms), and transmembrane transporters (ABCB1, ABCC1, ABCG2, SLCO1B1) make difficult the identification of patients with an optimal sensitivity and specificity, and a large part of variability among patients still remains unexplained. Irinotecan 41-51 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 198-203 21787264-6 2011 However, the complex pharmacokinetics of irinotecan and the involvement of several enzymes other than UGT (i.e., carboxyl estherases, CYP450 isoforms), and transmembrane transporters (ABCB1, ABCC1, ABCG2, SLCO1B1) make difficult the identification of patients with an optimal sensitivity and specificity, and a large part of variability among patients still remains unexplained. Irinotecan 41-51 solute carrier organic anion transporter family member 1B1 Homo sapiens 205-212 21652203-6 2011 Although exploratory nature and small sample size may be limitations of this study, these findings indicate that the efficacy of irinotecan plus cetuximab in MCRC patients with wild-type KRAS did not differ by previous sensitivity to irinotecan. Irinotecan 129-139 KRAS proto-oncogene, GTPase Homo sapiens 187-191 22077505-3 2011 METHODS: Published papers and publicly accessible genomic databases were searched up to August 2009 to obtain allele and genotype frequencies for UGT1A1*6 and *28, two common variants of UGT1A1, a gene that encodes for a key enzyme in the pathway of irinotecan metabolism. Irinotecan 250-260 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 146-152 22077505-3 2011 METHODS: Published papers and publicly accessible genomic databases were searched up to August 2009 to obtain allele and genotype frequencies for UGT1A1*6 and *28, two common variants of UGT1A1, a gene that encodes for a key enzyme in the pathway of irinotecan metabolism. Irinotecan 250-260 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 187-193 22077505-6 2011 Indians are at an elevated risk of irinotecan-induced neutropenia associated with UGT1A1*28 compared with Chinese and Japanese, and at an even higher risk compared with North American Caucasians. Irinotecan 35-45 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 82-88 22077505-7 2011 On the other hand, Chinese and Japanese are at an elevated risk of irinotecan-induced neutropenia associated with UGT1A1*6 relative to Indians in Singapore or North American Caucasians. Irinotecan 67-77 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 114-120 21744079-1 2011 We determined the maximum tolerated dose (MTD) and dose-limiting toxicities (DLT) of the oral vascular endothelial growth factor receptor (VEGFR) inhibitor, sunitinib, when administered with irinotecan among recurrent malignant glioma (MG) patients. Irinotecan 191-201 kinase insert domain receptor Homo sapiens 94-137 21744079-1 2011 We determined the maximum tolerated dose (MTD) and dose-limiting toxicities (DLT) of the oral vascular endothelial growth factor receptor (VEGFR) inhibitor, sunitinib, when administered with irinotecan among recurrent malignant glioma (MG) patients. Irinotecan 191-201 kinase insert domain receptor Homo sapiens 139-144 21997136-0 2011 An EGFR inhibitor enhances the efficacy of SN38, an active metabolite of irinotecan, in SN38-refractory gastric carcinoma cells. Irinotecan 73-83 epidermal growth factor receptor Homo sapiens 3-7 22333276-0 2011 [Study of irinotecan-induced toxicity and its correlation to UGT1A1 gene promoter polymorphisms]. Irinotecan 10-20 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 61-67 22333276-1 2011 OBJECTIVES: To investigate the distribution of uridine diphosphate-glucuronosyltransferase 1A1 (UGT1A1) gene promoter polymorphism and its relation to the toxicities caused by irinotecan in Chinese patients with cervical cancer and ovarian cancer. Irinotecan 176-186 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 47-94 22333276-1 2011 OBJECTIVES: To investigate the distribution of uridine diphosphate-glucuronosyltransferase 1A1 (UGT1A1) gene promoter polymorphism and its relation to the toxicities caused by irinotecan in Chinese patients with cervical cancer and ovarian cancer. Irinotecan 176-186 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 96-102 22333276-5 2011 The relationship between UGT1A1 gene promoter polymorphism and the toxicity caused by irinotecan was analyzed. Irinotecan 86-96 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 25-31 21990354-7 2011 Through the Nrf2-dependent increase of proteasomal gene expression and proteasome activity, the sensitivity of NCM460 cells to tumor necrosis factor-related apoptosis-inducing ligand- or irinotecan-induced apoptosis declined. Irinotecan 187-197 NFE2 like bZIP transcription factor 2 Homo sapiens 12-16 22044840-0 2011 Anticancer drug irinotecan inhibits homomeric 5-HT3A and heteromeric 5-HT3AB receptor responses. Irinotecan 16-26 5-hydroxytryptamine receptor 3A Homo sapiens 46-52 21997136-2 2011 This study was performed to clarify the effect of epidermal growth factor receptor (EGFR) inhibitors in combination with SN38, an active metabolite of irinotecan, on the proliferation of irinotecan-refractory gastric cancer. Irinotecan 187-197 epidermal growth factor receptor Homo sapiens 50-82 21997136-2 2011 This study was performed to clarify the effect of epidermal growth factor receptor (EGFR) inhibitors in combination with SN38, an active metabolite of irinotecan, on the proliferation of irinotecan-refractory gastric cancer. Irinotecan 187-197 epidermal growth factor receptor Homo sapiens 84-88 21997136-7 2011 The combination of an EGFR inhibitor and SN38 significantly increased the levels of apoptosis-related molecules, caspase-6, p53, and DAPK-2, and resulted in the induction of apoptosis of irinotecan-resistant cells. Irinotecan 187-197 epidermal growth factor receptor Homo sapiens 22-26 21997136-7 2011 The combination of an EGFR inhibitor and SN38 significantly increased the levels of apoptosis-related molecules, caspase-6, p53, and DAPK-2, and resulted in the induction of apoptosis of irinotecan-resistant cells. Irinotecan 187-197 death associated protein kinase 2 Homo sapiens 133-139 21997136-8 2011 The EGFR inhibitors increased the S-phase and decreased the UGT1A1 and ABCG expression in irinotecan-resistant cells. Irinotecan 90-100 epidermal growth factor receptor Homo sapiens 4-8 21997136-8 2011 The EGFR inhibitors increased the S-phase and decreased the UGT1A1 and ABCG expression in irinotecan-resistant cells. Irinotecan 90-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 60-66 21997136-10 2011 CONCLUSION: The combination treatment with an EGFR inhibitor and irinotecan might produce synergistic anti-tumour effects for irinotecan-refractory gastric cancer cells. Irinotecan 126-136 epidermal growth factor receptor Homo sapiens 46-50 21869821-3 2011 We genetically modified HB1.F3 (F3) immortalized human NSCs to express rabbit carboxylesterase (rCE) enzyme, which efficiently converts the prodrug CPT-11 (Irinotecan) into an active anti-cancer agent (SN-38). Irinotecan 148-154 coagulation factor III, tissue factor Homo sapiens 28-30 21892003-0 2011 Single nucleotide polymorphisms of ABCC5 and ABCG1 transporter genes correlate to irinotecan-associated gastrointestinal toxicity in colorectal cancer patients: a DMET microarray profiling study. Irinotecan 82-92 ATP binding cassette subfamily C member 5 Homo sapiens 35-40 21869821-3 2011 We genetically modified HB1.F3 (F3) immortalized human NSCs to express rabbit carboxylesterase (rCE) enzyme, which efficiently converts the prodrug CPT-11 (Irinotecan) into an active anti-cancer agent (SN-38). Irinotecan 156-166 coagulation factor III, tissue factor Homo sapiens 24-30 21892003-0 2011 Single nucleotide polymorphisms of ABCC5 and ABCG1 transporter genes correlate to irinotecan-associated gastrointestinal toxicity in colorectal cancer patients: a DMET microarray profiling study. Irinotecan 82-92 ATP binding cassette subfamily G member 1 Homo sapiens 45-50 21892003-1 2011 Recent findings have disclosed the role of UDP-glucuronosyltransferase (UGT) 1A1*28 on the haematological toxicity induced by irinotecan (CPT-11), a drug commonly used in the treatment of metastatic colorectal cancer (mCRC). Irinotecan 126-136 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 43-80 21892003-1 2011 Recent findings have disclosed the role of UDP-glucuronosyltransferase (UGT) 1A1*28 on the haematological toxicity induced by irinotecan (CPT-11), a drug commonly used in the treatment of metastatic colorectal cancer (mCRC). Irinotecan 138-144 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 43-80 21869821-3 2011 We genetically modified HB1.F3 (F3) immortalized human NSCs to express rabbit carboxylesterase (rCE) enzyme, which efficiently converts the prodrug CPT-11 (Irinotecan) into an active anti-cancer agent (SN-38). Irinotecan 148-154 coagulation factor III, tissue factor Homo sapiens 24-30 21869821-3 2011 We genetically modified HB1.F3 (F3) immortalized human NSCs to express rabbit carboxylesterase (rCE) enzyme, which efficiently converts the prodrug CPT-11 (Irinotecan) into an active anti-cancer agent (SN-38). Irinotecan 156-166 coagulation factor III, tissue factor Homo sapiens 28-30 21892003-10 2011 We have identified 3 SNPs mapping in ABCG1, ABCC5 and OATP1B1/SLCO1B1 transporter genes associated with GI toxicity induced by irinotecan in mCRC patients expanding the available knowledge of irinogenomics. Irinotecan 127-137 ATP binding cassette subfamily G member 1 Homo sapiens 37-42 21892003-10 2011 We have identified 3 SNPs mapping in ABCG1, ABCC5 and OATP1B1/SLCO1B1 transporter genes associated with GI toxicity induced by irinotecan in mCRC patients expanding the available knowledge of irinogenomics. Irinotecan 127-137 ATP binding cassette subfamily C member 5 Homo sapiens 44-49 21869821-3 2011 We genetically modified HB1.F3 (F3) immortalized human NSCs to express rabbit carboxylesterase (rCE) enzyme, which efficiently converts the prodrug CPT-11 (Irinotecan) into an active anti-cancer agent (SN-38). Irinotecan 202-207 coagulation factor III, tissue factor Homo sapiens 24-30 21892003-10 2011 We have identified 3 SNPs mapping in ABCG1, ABCC5 and OATP1B1/SLCO1B1 transporter genes associated with GI toxicity induced by irinotecan in mCRC patients expanding the available knowledge of irinogenomics. Irinotecan 127-137 solute carrier organic anion transporter family member 1B1 Homo sapiens 54-61 21869821-3 2011 We genetically modified HB1.F3 (F3) immortalized human NSCs to express rabbit carboxylesterase (rCE) enzyme, which efficiently converts the prodrug CPT-11 (Irinotecan) into an active anti-cancer agent (SN-38). Irinotecan 202-207 coagulation factor III, tissue factor Homo sapiens 28-30 21892003-10 2011 We have identified 3 SNPs mapping in ABCG1, ABCC5 and OATP1B1/SLCO1B1 transporter genes associated with GI toxicity induced by irinotecan in mCRC patients expanding the available knowledge of irinogenomics. Irinotecan 127-137 solute carrier organic anion transporter family member 1B1 Homo sapiens 62-69 21816982-7 2011 Moreover, MDCKII/BCRP/PDZK1 cells are more resistant than MDCKII/BCRP cells to the cytotoxicity of the anticancer agent 7-ethyl-10-hydroxycamptothecin (SN-38), which is a substrate of BCRP. Irinotecan 120-150 ATP binding cassette subfamily G member 2 Canis lupus familiaris 17-21 21816982-7 2011 Moreover, MDCKII/BCRP/PDZK1 cells are more resistant than MDCKII/BCRP cells to the cytotoxicity of the anticancer agent 7-ethyl-10-hydroxycamptothecin (SN-38), which is a substrate of BCRP. Irinotecan 120-150 Na(+)/H(+) exchange regulatory cofactor NHE-RF3 Canis lupus familiaris 22-27 21816982-7 2011 Moreover, MDCKII/BCRP/PDZK1 cells are more resistant than MDCKII/BCRP cells to the cytotoxicity of the anticancer agent 7-ethyl-10-hydroxycamptothecin (SN-38), which is a substrate of BCRP. Irinotecan 120-150 ATP binding cassette subfamily G member 2 Canis lupus familiaris 65-69 21816982-7 2011 Moreover, MDCKII/BCRP/PDZK1 cells are more resistant than MDCKII/BCRP cells to the cytotoxicity of the anticancer agent 7-ethyl-10-hydroxycamptothecin (SN-38), which is a substrate of BCRP. Irinotecan 120-150 ATP binding cassette subfamily G member 2 Canis lupus familiaris 65-69 21816982-7 2011 Moreover, MDCKII/BCRP/PDZK1 cells are more resistant than MDCKII/BCRP cells to the cytotoxicity of the anticancer agent 7-ethyl-10-hydroxycamptothecin (SN-38), which is a substrate of BCRP. Irinotecan 152-157 ATP binding cassette subfamily G member 2 Canis lupus familiaris 17-21 21816982-7 2011 Moreover, MDCKII/BCRP/PDZK1 cells are more resistant than MDCKII/BCRP cells to the cytotoxicity of the anticancer agent 7-ethyl-10-hydroxycamptothecin (SN-38), which is a substrate of BCRP. Irinotecan 152-157 Na(+)/H(+) exchange regulatory cofactor NHE-RF3 Canis lupus familiaris 22-27 21948564-8 2011 Surprisingly, expression of miR-451 caused a decrease in self-renewal, tumorigenicity, and chemoresistance to irinotecan of colonspheres. Irinotecan 110-120 microRNA 451a Homo sapiens 28-35 22045708-10 2011 SPECT analyses demonstrated a significant increase in tumoral (99m)Tc-(CO)(3) His-annexin A5 uptake 4 d after bevacizumab treatment and 24 h after irinotecan administration (232.78 +- 24.82 percentage injected dose/tumor weight [g]/body weight [kg], P < 0.05), compared with each monotherapy, indicating a synergistic effect of both therapies. Irinotecan 147-157 annexin A5 Mus musculus 82-92 22045708-11 2011 CONCLUSION: (99m)Tc-(CO)(3) His-annexin A5 micro-SPECT demonstrates increased antitumor activity of irinotecan during the transient vascular normalization period caused by bevacizumab. Irinotecan 100-110 annexin A5 Mus musculus 32-42 21529984-6 2011 The expression status of Klotho, and of the ATP-binding cassette (ABC) transporters MRP1, MDR and breast cancer resistant protein (BCRP), which can cause resistance to anticancer drugs, including irinotecan, was assessed by immunohistochemical analysis in resected surgical specimens of patients with early-stage SCLC. Irinotecan 196-206 ATP binding cassette subfamily C member 1 Homo sapiens 84-88 22045708-0 2011 (99)mTc-(CO)(3) His-annexin A5 micro-SPECT demonstrates increased cell death by irinotecan during the vascular normalization window caused by bevacizumab. Irinotecan 80-90 annexin A5 Mus musculus 20-30 21948564-12 2011 Furthermore, miR-451 restoration decreases expression of the ATP-binding cassette drug transporter ABCB1 and results in irinotecan sensitization. Irinotecan 120-130 microRNA 451a Homo sapiens 13-20 21948564-13 2011 These findings correlate well with the lower expression of miR-451 observed in patients who did not respond to irinotecan-based first-line therapy compared with patients who did. Irinotecan 111-121 microRNA 451a Homo sapiens 59-66 21948564-14 2011 Our data suggest that miR-451 is a novel candidate to circumvent recurrence and drug resistance in colorectal cancer and could be used as a marker to predict response to irinotecan in patients with colon carcinoma. Irinotecan 170-180 microRNA 451a Homo sapiens 22-29 21918342-14 2011 Cox regression analysis showed that irinotecan-containing regimens were associated with improved overall survival(hazard ratio: 0. Irinotecan 36-46 cytochrome c oxidase subunit 8A Homo sapiens 0-3 21210765-2 2011 Irinotecan and topotecan are currently used to treat various types of cancers and many CPT derivatives are being developed. Irinotecan 0-10 choline phosphotransferase 1 Homo sapiens 87-90 21210765-9 2011 Thus, the present study has identified RECQL5 as a major determinant for CPT resistance in colorectal cancer cells and a potential candidate as a biomarker for irinotecan-based treatment for colon cancer. Irinotecan 160-170 RecQ like helicase 5 Homo sapiens 39-45 21286718-0 2011 Cetuximab enhances the activities of irinotecan on gastric cancer cell lines through downregulating the EGFR pathway upregulated by irinotecan. Irinotecan 37-47 epidermal growth factor receptor Homo sapiens 104-108 21286718-0 2011 Cetuximab enhances the activities of irinotecan on gastric cancer cell lines through downregulating the EGFR pathway upregulated by irinotecan. Irinotecan 132-142 epidermal growth factor receptor Homo sapiens 104-108 21286718-6 2011 The effects of cetuximab or irinotecan as single agents or the combination on the expression of p53, p16, and EGFR signaling pathways were also studied. Irinotecan 28-38 tumor protein p53 Homo sapiens 96-99 21286718-6 2011 The effects of cetuximab or irinotecan as single agents or the combination on the expression of p53, p16, and EGFR signaling pathways were also studied. Irinotecan 28-38 cyclin dependent kinase inhibitor 2A Homo sapiens 101-104 21286718-6 2011 The effects of cetuximab or irinotecan as single agents or the combination on the expression of p53, p16, and EGFR signaling pathways were also studied. Irinotecan 28-38 epidermal growth factor receptor Homo sapiens 110-114 21286718-8 2011 Irinotecan increases phosphorylation of EGFR, MAPK, and AKT and decreases the expression of P27(Kip1), which could be all abrogated by its combination with cetuximab. Irinotecan 0-10 epidermal growth factor receptor Homo sapiens 40-44 21286718-8 2011 Irinotecan increases phosphorylation of EGFR, MAPK, and AKT and decreases the expression of P27(Kip1), which could be all abrogated by its combination with cetuximab. Irinotecan 0-10 AKT serine/threonine kinase 1 Homo sapiens 56-59 21286718-8 2011 Irinotecan increases phosphorylation of EGFR, MAPK, and AKT and decreases the expression of P27(Kip1), which could be all abrogated by its combination with cetuximab. Irinotecan 0-10 interferon alpha inducible protein 27 Homo sapiens 92-95 21286718-8 2011 Irinotecan increases phosphorylation of EGFR, MAPK, and AKT and decreases the expression of P27(Kip1), which could be all abrogated by its combination with cetuximab. Irinotecan 0-10 cyclin dependent kinase inhibitor 1B Homo sapiens 96-100 21286718-10 2011 CONCLUSIONS : Cetuximab enhances the activities of irinotecan on GC cells via the downregulation of the EGFR pathway upregulated by irinotecan. Irinotecan 100-110 epidermal growth factor receptor Homo sapiens 153-157 21286718-10 2011 CONCLUSIONS : Cetuximab enhances the activities of irinotecan on GC cells via the downregulation of the EGFR pathway upregulated by irinotecan. Irinotecan 181-191 epidermal growth factor receptor Homo sapiens 153-157 20467885-10 2011 Irinotecan and simvastatin combination treatment of A549 and H460 cells increased G(1) phase arrest, which was associated with up-regulation of p21(WAF1/CIP) and p53 compared with irinotecan alone. Irinotecan 0-10 cyclin dependent kinase inhibitor 1A Homo sapiens 144-147 20467885-10 2011 Irinotecan and simvastatin combination treatment of A549 and H460 cells increased G(1) phase arrest, which was associated with up-regulation of p21(WAF1/CIP) and p53 compared with irinotecan alone. Irinotecan 0-10 tumor protein p53 Homo sapiens 162-165 21850716-0 2011 Total synthesis of 7-ethyl-10-hydroxycamptothecin (SN38) and its application to the development of C18-functionalized camptothecin derivatives. Irinotecan 19-49 Bardet-Biedl syndrome 9 Homo sapiens 99-102 21868538-1 2011 The present study has investigated the effect of panipenem, a widely used antibiotic, on the pharmacokinetics of an active metabolite of irinotecan (CPT-11), 7-ethyl-10-hydroxy-camptothecin (SN-38) and SN-38 glucuronide (SN-38G) produced by uridine-diphosphate glucuronosyltransferase (UGT) 1A isoform-mediated glucuronidation in rats. Irinotecan 137-147 Ugt1a@ Rattus norvegicus 241-293 21868538-1 2011 The present study has investigated the effect of panipenem, a widely used antibiotic, on the pharmacokinetics of an active metabolite of irinotecan (CPT-11), 7-ethyl-10-hydroxy-camptothecin (SN-38) and SN-38 glucuronide (SN-38G) produced by uridine-diphosphate glucuronosyltransferase (UGT) 1A isoform-mediated glucuronidation in rats. Irinotecan 149-155 Ugt1a@ Rattus norvegicus 241-293 21868538-3 2011 When the effect of pretreatment with panipenem on glucuronidation activities of substrates for hepatic UGT1A isoforms was investigated using substrates 4-methylumbelliferone (4MU), estradiol and SN-38, the rate of 4MU glucuronide formation was significantly increased, but that of estradiol glucuronide formation was unchanged. Irinotecan 195-200 Ugt1a@ Rattus norvegicus 103-108 21855038-1 2011 BACKGROUND: This prospective analysis evaluated the effect of tumor KRAS status on efficacy of second-line panitumumab plus folinic acid/5-fluorouracil/irinotecan (FOLFIRI). Irinotecan 152-162 KRAS proto-oncogene, GTPase Homo sapiens 68-72 21945893-14 2011 Cox regression analysis showed that irinotecan-containing regimens were associated with improved overall survival(hazard ratio: 0. Irinotecan 36-46 cytochrome c oxidase subunit 8A Homo sapiens 0-3 21945437-3 2011 As an early response to DNA damage following CPT-11 treatment, we found that there was an increase in the gammaH2AX(S139) foci number and that total phosphorylation levels were reduced in PC-3 cells following ectopic NKX3.1 expression as well as in LNCaP cells following androgen administration. Irinotecan 45-51 NK3 homeobox 1 Homo sapiens 217-223 21965746-0 2011 ERas enhances resistance to CPT-11 in gastric cancer. Irinotecan 28-34 ES cell expressed Ras Homo sapiens 0-4 21965746-5 2011 RESULTS: ERas-overexpressing clones were significantly more resistant to CPT-11 than were the control (p<0.001). Irinotecan 73-79 ES cell expressed Ras Homo sapiens 9-13 21965746-9 2011 CONCLUSION: ERas induces chemoresistance to CPT-11 via activation of phosphatidylinositol-3 kinase-protein kinase beta mTOR pathway and NF-kappaB, and consequently results in up-regulation of ABCG2. Irinotecan 44-50 ES cell expressed Ras Homo sapiens 12-16 21965746-9 2011 CONCLUSION: ERas induces chemoresistance to CPT-11 via activation of phosphatidylinositol-3 kinase-protein kinase beta mTOR pathway and NF-kappaB, and consequently results in up-regulation of ABCG2. Irinotecan 44-50 mechanistic target of rapamycin kinase Homo sapiens 119-123 21965746-9 2011 CONCLUSION: ERas induces chemoresistance to CPT-11 via activation of phosphatidylinositol-3 kinase-protein kinase beta mTOR pathway and NF-kappaB, and consequently results in up-regulation of ABCG2. Irinotecan 44-50 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 192-197 21740478-0 2011 Genotype-directed, dose-finding study of irinotecan in cancer patients with UGT1A1*28 and/or UGT1A1*6 polymorphisms. Irinotecan 41-51 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 76-82 21740478-0 2011 Genotype-directed, dose-finding study of irinotecan in cancer patients with UGT1A1*28 and/or UGT1A1*6 polymorphisms. Irinotecan 41-51 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-99 21740478-1 2011 Irinotecan-induced severe neutropenia is associated with homozygosity for the UGT1A1*28 or UGT1A1*6 alleles. Irinotecan 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 78-84 21740478-1 2011 Irinotecan-induced severe neutropenia is associated with homozygosity for the UGT1A1*28 or UGT1A1*6 alleles. Irinotecan 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 91-97 21740478-2 2011 In this study, we determined the maximum-tolerated dose (MTD) of irinotecan in patients with UGT1A1 polymorphisms. Irinotecan 65-75 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-99 21740478-14 2011 Patients homozygous for the UGT1A1*28 or UGT1A1*6 allele can receive irinotecan in a starting dose of 150 mg/m(2), but many required dose reductions or delayed treatment in subsequent cycles. Irinotecan 69-79 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 28-34 21740478-14 2011 Patients homozygous for the UGT1A1*28 or UGT1A1*6 allele can receive irinotecan in a starting dose of 150 mg/m(2), but many required dose reductions or delayed treatment in subsequent cycles. Irinotecan 69-79 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 41-47 21452059-8 2011 Further, western blot analyses of xenograft tumors demonstrated that EZN-2208 had significantly more effect than CPT-11 in down-regulating HIF-1alpha, VEGF, Glut1 and MMP2 protein levels. Irinotecan 113-119 hypoxia inducible factor 1, alpha subunit Mus musculus 139-149 21452059-8 2011 Further, western blot analyses of xenograft tumors demonstrated that EZN-2208 had significantly more effect than CPT-11 in down-regulating HIF-1alpha, VEGF, Glut1 and MMP2 protein levels. Irinotecan 113-119 vascular endothelial growth factor A Mus musculus 151-155 21452059-8 2011 Further, western blot analyses of xenograft tumors demonstrated that EZN-2208 had significantly more effect than CPT-11 in down-regulating HIF-1alpha, VEGF, Glut1 and MMP2 protein levels. Irinotecan 113-119 solute carrier family 2 (facilitated glucose transporter), member 1 Mus musculus 157-162 21452059-8 2011 Further, western blot analyses of xenograft tumors demonstrated that EZN-2208 had significantly more effect than CPT-11 in down-regulating HIF-1alpha, VEGF, Glut1 and MMP2 protein levels. Irinotecan 113-119 matrix metallopeptidase 2 Mus musculus 167-171 21771946-2 2011 The UGT1A1 promoter genotype also correlates with toxicity induced by the chemotherapeutic drug irinotecan. Irinotecan 96-106 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 22866151-0 2011 Prevalence of topoisomerase I genetic mutations and UGT1A1 polymorphisms associated with irinotecan in individuals of Asian descent. Irinotecan 89-99 DNA topoisomerase I Homo sapiens 14-29 21687948-4 2011 Polymorphisms of MRP2, a protein involved in methotrexate, cisplatin and irinotecan active metabolite glucuronide transport, negatively affect platinum-based chemotherapy response. Irinotecan 73-83 ATP binding cassette subfamily C member 2 Homo sapiens 17-21 22866151-0 2011 Prevalence of topoisomerase I genetic mutations and UGT1A1 polymorphisms associated with irinotecan in individuals of Asian descent. Irinotecan 89-99 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 52-58 22866151-1 2011 Topoisomerase I (TOP-I) mutations have been shown to be correlated to irinotecan resistance in vitro. Irinotecan 70-80 DNA topoisomerase I Homo sapiens 0-15 22866151-1 2011 Topoisomerase I (TOP-I) mutations have been shown to be correlated to irinotecan resistance in vitro. Irinotecan 70-80 DNA topoisomerase I Homo sapiens 17-22 22866151-3 2011 On the other hand, polymorphisms of UGT1A1 have been shown to be associated with CPT-11 toxicity in clinical situations. Irinotecan 81-87 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 36-42 22866151-4 2011 The primary aim of this study was to investigate the prevalence of mutations in the TOP-I exons associated with CPT-11 resistance, including untreated cancer tissue. Irinotecan 112-118 DNA topoisomerase I Homo sapiens 84-89 22866151-12 2011 In conclusion, TOP-I genetic mutations correlated to CPT-11 resistance were not detected in any of the subjects and untreated lung cancer tissues. Irinotecan 53-59 DNA topoisomerase I Homo sapiens 15-20 21829065-3 2011 In this study, we tested the point mutation of codon 12 and 13 in the KRAS gene by Luminex(xMAP)flow cytometry with sequence-specific oligonucleotide probes for 39 out of 64 unresectable mCRC patients enrolled from Sep 2008 to Oct 2009, who were administered cetuximab in combination with irinotecan(CPT-11)as third-line therapy. Irinotecan 289-299 KRAS proto-oncogene, GTPase Homo sapiens 70-74 21046105-10 2011 [(18)F]FLT-PET imaging revealed a 64% decrease in [(18)F]FLT uptake in tumors of HCT116 xenograft-bearing mice treated with a combination of PXD101 and irinotecan, indicating a decrease in thymidine kinase 1 (TK1) activity. Irinotecan 152-162 thymidine kinase 1 Mus musculus 189-207 21046105-10 2011 [(18)F]FLT-PET imaging revealed a 64% decrease in [(18)F]FLT uptake in tumors of HCT116 xenograft-bearing mice treated with a combination of PXD101 and irinotecan, indicating a decrease in thymidine kinase 1 (TK1) activity. Irinotecan 152-162 thymidine kinase 1 Mus musculus 209-212 21829065-3 2011 In this study, we tested the point mutation of codon 12 and 13 in the KRAS gene by Luminex(xMAP)flow cytometry with sequence-specific oligonucleotide probes for 39 out of 64 unresectable mCRC patients enrolled from Sep 2008 to Oct 2009, who were administered cetuximab in combination with irinotecan(CPT-11)as third-line therapy. Irinotecan 300-306 KRAS proto-oncogene, GTPase Homo sapiens 70-74 21829065-33 2011 Therefore, we concluded that the KRAS mutation in mCRC is a predictive factor for the lack of response to combination therapy with cetuximab plus CPT- 11, as reported in previous clinical studies. Irinotecan 146-153 KRAS proto-oncogene, GTPase Homo sapiens 33-37 20072801-0 2011 Phase II study of combination chemotherapy with irinotecan and cetuximab for pretreated metastatic colorectal cancer harboring wild-type KRAS. Irinotecan 48-58 KRAS proto-oncogene, GTPase Homo sapiens 137-141 20072801-1 2011 The aim of this study was to prospectively evaluate the efficacy of combination irinotecan and cetuximab chemotherapy in patients with pretreated metastatic colorectal cancer harboring wild-type KRAS. Irinotecan 80-90 KRAS proto-oncogene, GTPase Homo sapiens 195-199 21287524-2 2011 The UGT1A1 gene encodes for an enzyme that metabolizes irinotecan, and its genetic variants were shown to be associated with increased drug toxicity. Irinotecan 55-65 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 21585235-6 2011 This achievement has led to personalized irinotecan therapy, reflecting ethnic differences in UGT1A1 genotypes, and possible benefits of genetic testing have also been suggested for gemcitabine and tamoxifen therapy, which still requires further validation. Irinotecan 41-51 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 94-100 21868346-1 2011 OBJECTIVE: To evaluate the safety and efficacy of trsatuzumab (Herceptin) combined with FOLFIRI regimen (irinotecan plus 5-FU/LV) in the treatment of HER2-positive advanced gastric cancer. Irinotecan 105-115 erb-b2 receptor tyrosine kinase 2 Homo sapiens 150-154 21287524-12 2011 CONCLUSIONS: The UGT1A1*28 7/7 genotype is strongly associated with severe hematological toxicity and higher hospitalization rate and predicts lower survival of colorectal cancer in users of irinotecan. Irinotecan 191-201 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 17-23 21733184-0 2011 Irinotecan induces steroid and xenobiotic receptor (SXR) signaling to detoxification pathway in colon cancer cells. Irinotecan 0-10 nuclear receptor subfamily 1 group I member 2 Homo sapiens 19-50 21733184-0 2011 Irinotecan induces steroid and xenobiotic receptor (SXR) signaling to detoxification pathway in colon cancer cells. Irinotecan 0-10 nuclear receptor subfamily 1 group I member 2 Homo sapiens 52-55 21733184-5 2011 RESULTS: In this study, we have shown that endogenous SXR is activated in response to SN-38, the active metabolite of the anticancer drug irinotecan, in human colon cancer cell lines. Irinotecan 86-91 nuclear receptor subfamily 1 group I member 2 Homo sapiens 54-57 21733184-5 2011 RESULTS: In this study, we have shown that endogenous SXR is activated in response to SN-38, the active metabolite of the anticancer drug irinotecan, in human colon cancer cell lines. Irinotecan 138-148 nuclear receptor subfamily 1 group I member 2 Homo sapiens 54-57 21733184-6 2011 We have found that endogenous SXR translocates into the nucleus and associates with RXR upon SN-38 treatment. Irinotecan 93-98 nuclear receptor subfamily 1 group I member 2 Homo sapiens 30-33 21733184-6 2011 We have found that endogenous SXR translocates into the nucleus and associates with RXR upon SN-38 treatment. Irinotecan 93-98 retinoid X receptor alpha Homo sapiens 84-87 21733184-9 2011 As a consequence, cells overexpressing SXR were found to be less sensitive to irinotecan treatment. Irinotecan 78-88 nuclear receptor subfamily 1 group I member 2 Homo sapiens 39-42 21733184-10 2011 CONCLUSIONS: Altogether, these results suggest that the SXR pathway is involved in colon cancer irinotecan resistance in colon cancer cell line via the upregulation of select detoxification genes. Irinotecan 96-106 nuclear receptor subfamily 1 group I member 2 Homo sapiens 56-59 21459094-10 2011 Inhibition of the drug-efflux pump ABCB1 by PSC-833 allowed irinotecan to eradicate tumor-initiating cells. Irinotecan 60-70 ATP binding cassette subfamily B member 1 Homo sapiens 35-40 21459094-15 2011 CONCLUSIONS: The resistance of colorectal tumors to irinotecan requires the cooperative action of tumor-initiating ALDHhigh/ABCB1negative cells and their differentiated, drug-expelling, ALDHlow/ABCB1positive daughter cells. Irinotecan 52-62 ATP binding cassette subfamily B member 1 Homo sapiens 124-129 21459094-15 2011 CONCLUSIONS: The resistance of colorectal tumors to irinotecan requires the cooperative action of tumor-initiating ALDHhigh/ABCB1negative cells and their differentiated, drug-expelling, ALDHlow/ABCB1positive daughter cells. Irinotecan 52-62 ATP binding cassette subfamily B member 1 Homo sapiens 194-199 21464197-6 2011 FLB-12 significantly inhibits the PXR-activated loss of righting reflex to 2,2,2-tribromoethanol (Avertin) in vivo, abrogates PXR-mediated resistance to 7-ethyl-10-hydroxycamptothecin (SN-38) in colon cancer cells in vitro, and attenuates PXR-mediated acetaminophen hepatotoxicity in vivo. Irinotecan 153-183 nuclear receptor subfamily 1, group I, member 2 Mus musculus 34-37 21566063-8 2011 Coadministration of irinotecan with YHO-13351 significantly increased the survival time of mice inoculated with BCRP-transduced murine leukemia P388 cells and suppressed the tumor growth in an HCT116/BCRP xenograft model, whereas irinotecan alone had little effect in these tumor models. Irinotecan 20-30 ATP binding cassette subfamily G member 2 (Junior blood group) Mus musculus 112-116 21566063-8 2011 Coadministration of irinotecan with YHO-13351 significantly increased the survival time of mice inoculated with BCRP-transduced murine leukemia P388 cells and suppressed the tumor growth in an HCT116/BCRP xenograft model, whereas irinotecan alone had little effect in these tumor models. Irinotecan 20-30 ATP binding cassette subfamily G member 2 (Junior blood group) Mus musculus 193-204 21566063-8 2011 Coadministration of irinotecan with YHO-13351 significantly increased the survival time of mice inoculated with BCRP-transduced murine leukemia P388 cells and suppressed the tumor growth in an HCT116/BCRP xenograft model, whereas irinotecan alone had little effect in these tumor models. Irinotecan 230-240 ATP binding cassette subfamily G member 2 (Junior blood group) Mus musculus 112-116 21566063-1 2011 Breast cancer resistance protein (BCRP/ABCG2) confers resistance to anticancer drugs such as 7-ethyl-10-hydroxycamptothecin (SN-38, an active metabolite of irinotecan), mitoxantrone, and topotecan. Irinotecan 93-123 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 0-32 21566063-1 2011 Breast cancer resistance protein (BCRP/ABCG2) confers resistance to anticancer drugs such as 7-ethyl-10-hydroxycamptothecin (SN-38, an active metabolite of irinotecan), mitoxantrone, and topotecan. Irinotecan 93-123 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 34-38 21566063-1 2011 Breast cancer resistance protein (BCRP/ABCG2) confers resistance to anticancer drugs such as 7-ethyl-10-hydroxycamptothecin (SN-38, an active metabolite of irinotecan), mitoxantrone, and topotecan. Irinotecan 93-123 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 39-44 21566063-1 2011 Breast cancer resistance protein (BCRP/ABCG2) confers resistance to anticancer drugs such as 7-ethyl-10-hydroxycamptothecin (SN-38, an active metabolite of irinotecan), mitoxantrone, and topotecan. Irinotecan 125-130 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 0-32 21566063-1 2011 Breast cancer resistance protein (BCRP/ABCG2) confers resistance to anticancer drugs such as 7-ethyl-10-hydroxycamptothecin (SN-38, an active metabolite of irinotecan), mitoxantrone, and topotecan. Irinotecan 125-130 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 34-38 21566063-1 2011 Breast cancer resistance protein (BCRP/ABCG2) confers resistance to anticancer drugs such as 7-ethyl-10-hydroxycamptothecin (SN-38, an active metabolite of irinotecan), mitoxantrone, and topotecan. Irinotecan 125-130 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 39-44 21566063-1 2011 Breast cancer resistance protein (BCRP/ABCG2) confers resistance to anticancer drugs such as 7-ethyl-10-hydroxycamptothecin (SN-38, an active metabolite of irinotecan), mitoxantrone, and topotecan. Irinotecan 156-166 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 0-32 21464197-6 2011 FLB-12 significantly inhibits the PXR-activated loss of righting reflex to 2,2,2-tribromoethanol (Avertin) in vivo, abrogates PXR-mediated resistance to 7-ethyl-10-hydroxycamptothecin (SN-38) in colon cancer cells in vitro, and attenuates PXR-mediated acetaminophen hepatotoxicity in vivo. Irinotecan 153-183 nuclear receptor subfamily 1, group I, member 2 Mus musculus 126-129 21566063-1 2011 Breast cancer resistance protein (BCRP/ABCG2) confers resistance to anticancer drugs such as 7-ethyl-10-hydroxycamptothecin (SN-38, an active metabolite of irinotecan), mitoxantrone, and topotecan. Irinotecan 156-166 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 34-38 21566063-1 2011 Breast cancer resistance protein (BCRP/ABCG2) confers resistance to anticancer drugs such as 7-ethyl-10-hydroxycamptothecin (SN-38, an active metabolite of irinotecan), mitoxantrone, and topotecan. Irinotecan 156-166 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 39-44 21566063-3 2011 YHO-13177 potentiated the cytotoxicity of SN-38, mitoxantrone, and topotecan in both BCRP-transduced human colon cancer HCT116 (HCT116/BCRP) cells and SN-38-resistant human lung cancer A549 (A549/SN4) cells that express BCRP, but had little effect in the parental cells. Irinotecan 42-47 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 85-89 21566063-4 2011 In addition, YHO-13177 potentiated the cytotoxicity of SN-38 in human lung cancer NCI-H460 and NCI-H23, myeloma RPMI-8226, and pancreatic cancer AsPC-1 cells that intrinsically expressed BCRP. Irinotecan 55-60 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 187-191 21464197-6 2011 FLB-12 significantly inhibits the PXR-activated loss of righting reflex to 2,2,2-tribromoethanol (Avertin) in vivo, abrogates PXR-mediated resistance to 7-ethyl-10-hydroxycamptothecin (SN-38) in colon cancer cells in vitro, and attenuates PXR-mediated acetaminophen hepatotoxicity in vivo. Irinotecan 153-183 nuclear receptor subfamily 1, group I, member 2 Mus musculus 126-129 21464197-6 2011 FLB-12 significantly inhibits the PXR-activated loss of righting reflex to 2,2,2-tribromoethanol (Avertin) in vivo, abrogates PXR-mediated resistance to 7-ethyl-10-hydroxycamptothecin (SN-38) in colon cancer cells in vitro, and attenuates PXR-mediated acetaminophen hepatotoxicity in vivo. Irinotecan 185-190 nuclear receptor subfamily 1, group I, member 2 Mus musculus 34-37 21654688-2 2011 Irinotecan is converted into 7-ethyl-10-hydroxycamptothecin (SN-38) by a carboxylsterase and metabolised through uridine diphosphate glucuronosyl transferase (UGT1A1). Irinotecan 29-59 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 159-165 21576795-3 2011 Therefore, we used the poly(NIPA-co-DMAEMA) nanoparticles as a carrier for the controlled release of a hydrophobic anticancer agent, 7-ethyl-10-hydroxy-camptothecin (SN-38). Irinotecan 166-171 zinc finger C3HC-type containing 1 Homo sapiens 28-32 21576795-6 2011 The cytotoxicity of the SN-38-loaded poly(NIPA-co-DMAEMA) nanoparticles was investigated in human colon cancer cells (HT-29) to compare with the treatment of an anticancer drug, Irinotecan( ) (CPT-11). Irinotecan 24-29 zinc finger C3HC-type containing 1 Homo sapiens 42-46 21654688-2 2011 Irinotecan is converted into 7-ethyl-10-hydroxycamptothecin (SN-38) by a carboxylsterase and metabolised through uridine diphosphate glucuronosyl transferase (UGT1A1). Irinotecan 61-66 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 159-165 21654688-2 2011 Irinotecan is converted into 7-ethyl-10-hydroxycamptothecin (SN-38) by a carboxylsterase and metabolised through uridine diphosphate glucuronosyl transferase (UGT1A1). Irinotecan 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 159-165 21486216-4 2011 We show that ONCOFID -S exerts a strong in vitro anti-proliferative activity on CD44 over-expressing rat DHD/K12/trb colon adenocarcinoma cells, as well as on gastric, breast, oesophageal, ovarian and lung human cancer cells, higher than that exerted by unconjugated SN-38. Irinotecan 267-272 CD44 molecule (Indian blood group) Rattus norvegicus 80-84 21350582-0 2011 Enhancement of CPT-11 antitumor activity by adenovirus-mediated expression of beta-glucuronidase in tumors. Irinotecan 15-21 glucuronidase beta Homo sapiens 78-96 21350582-3 2011 Here, we investigated if local expression of beta-glucuronidase (betaG) on cancer cells to catalytically convert SN38G to SN38 could enhance the antitumor activity of CPT-11. Irinotecan 167-173 glucuronidase beta Homo sapiens 45-63 21350582-5 2011 Surface beta-glucuronidase-expressing cells were 20 to 80-fold more sensitive to SN-38G than the parental cells. Irinotecan 81-86 glucuronidase beta Homo sapiens 8-26 21350582-6 2011 Intravenous CPT-11 produced significantly greater suppression of CL1-5 and LS174 T tumors that expressed betaG as compared with unmodified tumors. Irinotecan 12-18 adhesion G protein-coupled receptor L1 Homo sapiens 65-70 21559018-2 2011 The primary aim of our study was to verify a possible correlation between EGFR gene promoter methylation and clinical outcome in metastatic colorectal cancer patients receiving chemotherapy with irinotecan and cetuximab. Irinotecan 195-205 epidermal growth factor receptor Homo sapiens 74-78 20949523-11 2011 CONCLUSIONS: Tumor tissue lines established via subrenal capsule xenografting provide models with clinical relevance and the present study suggests that irinotecan could be useful for therapy of refractory prostatic SCC, in particular in combination with cisplatin. Irinotecan 153-163 serpin family B member 3 Homo sapiens 216-219 21372224-0 2011 Humanized anti-Trop-2 IgG-SN-38 conjugate for effective treatment of diverse epithelial cancers: preclinical studies in human cancer xenograft models and monkeys. Irinotecan 26-31 tumor associated calcium signal transducer 2 Homo sapiens 15-21 21372224-1 2011 PURPOSE: Evaluate the efficacy of an SN-38-anti-Trop-2 antibody-drug conjugate (ADC) against several human solid tumor types, and to assess its tolerability in mice and monkeys, the latter with tissue cross-reactivity to hRS7 similar to humans. Irinotecan 37-42 tumor associated calcium signal transducer 2 Homo sapiens 48-54 21372224-2 2011 EXPERIMENTAL DESIGN: Two SN-38 derivatives, CL2-SN-38 and CL2A-SN-38, were conjugated to the anti-Trop-2-humanized antibody, hRS7. Irinotecan 25-30 endogenous retrovirus group W member 5 Homo sapiens 44-47 21372224-2 2011 EXPERIMENTAL DESIGN: Two SN-38 derivatives, CL2-SN-38 and CL2A-SN-38, were conjugated to the anti-Trop-2-humanized antibody, hRS7. Irinotecan 25-30 tumor associated calcium signal transducer 2 Homo sapiens 98-104 21372224-7 2011 Exposure of cells to the ADC demonstrated signaling pathways leading to PARP cleavage, but differences versus free SN-38 in p53 and p21 upregulation were noted. Irinotecan 115-120 tumor protein p53 Homo sapiens 124-127 21372224-7 2011 Exposure of cells to the ADC demonstrated signaling pathways leading to PARP cleavage, but differences versus free SN-38 in p53 and p21 upregulation were noted. Irinotecan 115-120 H3 histone pseudogene 16 Homo sapiens 132-135 21372224-9 2011 Mice tolerated a dose of 2 x 12 mg/kg (SN-38 equivalents) with only short-lived elevations in ALT and AST liver enzyme levels. Irinotecan 39-44 transmembrane protease, serine 11d Mus musculus 102-105 21372224-11 2011 CONCLUSIONS: The anti-Trop-2 hRS7-CL2A-SN-38 ADC provides significant and specific antitumor effects against a range of human solid tumor types. Irinotecan 39-44 tumor associated calcium signal transducer 2 Homo sapiens 22-28 21721861-11 2011 However, the average expression of Rev-erbalpha, Per2, and Bmal1 were down-regulated 2- to 10-fold with irinotecan at the worst ZT, while being minimally or unaffected at the best ZT, irrespective of sex. Irinotecan 104-114 nuclear receptor subfamily 1, group D, member 1 Mus musculus 35-47 21721861-11 2011 However, the average expression of Rev-erbalpha, Per2, and Bmal1 were down-regulated 2- to 10-fold with irinotecan at the worst ZT, while being minimally or unaffected at the best ZT, irrespective of sex. Irinotecan 104-114 period circadian clock 2 Mus musculus 49-53 21721861-11 2011 However, the average expression of Rev-erbalpha, Per2, and Bmal1 were down-regulated 2- to 10-fold with irinotecan at the worst ZT, while being minimally or unaffected at the best ZT, irrespective of sex. Irinotecan 104-114 aryl hydrocarbon receptor nuclear translocator-like Mus musculus 59-64 21721861-13 2011 The mRNA expressions of irinotecan clock-controlled metabolism genes Ce2, Ugt1a1, and Top1 were unchanged or down-regulated according to irinotecan timing and sex. Irinotecan 24-34 UDP glucuronosyltransferase 1 family, polypeptide A1 Mus musculus 74-80 21721861-13 2011 The mRNA expressions of irinotecan clock-controlled metabolism genes Ce2, Ugt1a1, and Top1 were unchanged or down-regulated according to irinotecan timing and sex. Irinotecan 137-147 UDP glucuronosyltransferase 1 family, polypeptide A1 Mus musculus 74-80 21507240-8 2011 Specific depletion of Mcl-1 from HepG2 cells by RNA interference increases sensitivity of HepG2 cells to chemotherapeutic drugs (i.e. cisplatin and irinotecan). Irinotecan 148-158 MCL1 apoptosis regulator, BCL2 family member Homo sapiens 22-27 21387285-12 2011 In conclusion, administration of PDGF-R tyrosine kinase inhibitor in combination with irinotecan appears to impair the progressive growth of gastric carcinoma by blockade of PDGF-R signaling pathways in stromal cells. Irinotecan 86-96 platelet derived growth factor receptor, beta polypeptide Mus musculus 174-180 21298326-7 2011 Furthermore, the CD26 effect was enhanced when apigenin was paired with chemotherapeutic agents utilized in the treatment of advanced colorectal cancer including irinotecan, 5-fluorouracil and oxaliplatin. Irinotecan 162-172 dipeptidyl peptidase 4 Homo sapiens 17-21 21166471-8 2011 These findings indicated that Oct-targeted liposomes loaded with CPT-11 may offer considerable potential for MTC chemotherapy because cytotoxicity of both CPT-11 and Oct was enhanced by effective cellular uptake via SSTR2. Irinotecan 65-71 somatostatin receptor 2 Homo sapiens 216-221 21166471-8 2011 These findings indicated that Oct-targeted liposomes loaded with CPT-11 may offer considerable potential for MTC chemotherapy because cytotoxicity of both CPT-11 and Oct was enhanced by effective cellular uptake via SSTR2. Irinotecan 155-161 somatostatin receptor 2 Homo sapiens 216-221 21216137-2 2011 In vitro studies have shown that omeprazole can alter the function of metabolic enzymes and transporters that are involved in the metabolism of irinotecan, such as uridine diphosphate glucuronosyltransferase subfamily 1A1 (UGT1A1), cytochrome P-450 enzymes subfamily 3A (CYP3A) and ATP-binding cassette drug-transporter G2 (ABCG2). Irinotecan 144-154 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 223-229 21635994-6 2011 RESULTS: For the metastatic colorectal cancer LLD population with K-ras wild-type genotype, mean overall survival estimates were 37.7 months for first-line treatment with cetuximab plus FOLFIRI (irinotecan, leucovorin, fluorouracil) and 30.4 months for bevacizumab plus FOLFOX (oxaliplatin, leucovorin, fluorouracil). Irinotecan 195-205 KRAS proto-oncogene, GTPase Homo sapiens 66-71 21216137-2 2011 In vitro studies have shown that omeprazole can alter the function of metabolic enzymes and transporters that are involved in the metabolism of irinotecan, such as uridine diphosphate glucuronosyltransferase subfamily 1A1 (UGT1A1), cytochrome P-450 enzymes subfamily 3A (CYP3A) and ATP-binding cassette drug-transporter G2 (ABCG2). Irinotecan 144-154 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 282-322 21216137-2 2011 In vitro studies have shown that omeprazole can alter the function of metabolic enzymes and transporters that are involved in the metabolism of irinotecan, such as uridine diphosphate glucuronosyltransferase subfamily 1A1 (UGT1A1), cytochrome P-450 enzymes subfamily 3A (CYP3A) and ATP-binding cassette drug-transporter G2 (ABCG2). Irinotecan 144-154 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 324-329 21499002-7 2011 The patient was started on combined chemotherapy with S-1/CPT-11(S-1 80 mg/body, po, day 1-14 and CPT-11 120 mg/body iv day 1), serum CEA levels returned to normal range, and marked reduction of lymph node size was observed on CT. Irinotecan 58-64 proteasome 26S subunit, non-ATPase 1 Homo sapiens 65-68 21499002-7 2011 The patient was started on combined chemotherapy with S-1/CPT-11(S-1 80 mg/body, po, day 1-14 and CPT-11 120 mg/body iv day 1), serum CEA levels returned to normal range, and marked reduction of lymph node size was observed on CT. Irinotecan 58-64 CEA cell adhesion molecule 3 Homo sapiens 134-137 21242184-3 2011 Although she achieved long-term survival of more than 2 years, the last-line chemotherapy, consisting of irinotecan, induced grade 4 febrile neutropenia and her performance status deteriorated even with a reduced dose according to the analysis of UDP-glucuronosyltransferase 1A1. Irinotecan 105-115 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 247-278 21366639-4 2011 SN-38 is subsequently detoxified by uridine diphosphate-glycosyltransferase 1, encoded by the UGT1A1 gene. Irinotecan 0-5 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 94-100 21366639-5 2011 It is well known that the variant allele UGT1A*18 is associated with the more common adverse effects of irinotecan. Irinotecan 104-114 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 41-46 21104178-0 2011 Serum matrilysin correlates with poor survival independently of KRAS and BRAF status in refractory advanced colorectal cancer patients treated with irinotecan plus cetuximab. Irinotecan 148-158 matrix metallopeptidase 7 Homo sapiens 6-16 22214020-0 2011 [Treatment with CPT-11 based on the UGT1A1 genetic polymorphism]. Irinotecan 16-22 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 36-42 21439039-0 2011 EGFR related mutational status and association to clinical outcome of third-line cetuximab-irinotecan in metastatic colorectal cancer. Irinotecan 91-101 epidermal growth factor receptor Homo sapiens 0-4 21468552-5 2011 It has been demonstrated that the UGT1A1*28 polymorphism plays a predictive role in patients administered an irinotecan-containing regimen. Irinotecan 109-119 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 34-40 21309756-1 2011 Variation of a short (TA)(n) repeat sequence (rs8175347) covering the TATA box of UGT1A1 (UDP-glucuronosyltransferase1A1) is associated with hyperbilirubinaemia (Gilbert"s syndrome) and adverse drug reactions, and is used for dosage advice for irinotecan. Irinotecan 244-254 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 82-88 21309756-1 2011 Variation of a short (TA)(n) repeat sequence (rs8175347) covering the TATA box of UGT1A1 (UDP-glucuronosyltransferase1A1) is associated with hyperbilirubinaemia (Gilbert"s syndrome) and adverse drug reactions, and is used for dosage advice for irinotecan. Irinotecan 244-254 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 90-120 21193546-6 2011 In contrast, anti-DLL4 was efficacious against both wild-type and mutant KRAS colon tumors as a single agent and in combination with irinotecan. Irinotecan 133-143 delta like canonical Notch ligand 4 Homo sapiens 18-22 21193546-7 2011 Further analysis of mutant KRAS tumors indicated that the anti-DLL4/irinotecan combination produced a significant decrease in colon cancer stem cell frequency while promoting apoptosis in tumor cells. Irinotecan 68-78 KRAS proto-oncogene, GTPase Homo sapiens 27-31 21193546-7 2011 Further analysis of mutant KRAS tumors indicated that the anti-DLL4/irinotecan combination produced a significant decrease in colon cancer stem cell frequency while promoting apoptosis in tumor cells. Irinotecan 68-78 delta like canonical Notch ligand 4 Homo sapiens 63-67 21216589-1 2011 The breast cancer resistance protein ABCG2 confers cellular resistance to irinotecan (CPT-11) and its active metabolite SN-38. Irinotecan 74-84 ATP binding cassette subfamily G member 2 (Junior blood group) Mus musculus 37-42 21216589-1 2011 The breast cancer resistance protein ABCG2 confers cellular resistance to irinotecan (CPT-11) and its active metabolite SN-38. Irinotecan 86-92 ATP binding cassette subfamily G member 2 (Junior blood group) Mus musculus 37-42 21216589-1 2011 The breast cancer resistance protein ABCG2 confers cellular resistance to irinotecan (CPT-11) and its active metabolite SN-38. Irinotecan 120-125 ATP binding cassette subfamily G member 2 (Junior blood group) Mus musculus 37-42 21216589-4 2011 Addition of MBLI-87, an acridone derivative inhibitor, significantly increased the irinotecan effect against the growth of ABCG2-expressing xenografts. Irinotecan 83-93 ATP binding cassette subfamily G member 2 (Junior blood group) Mus musculus 123-128 21216589-5 2011 In vitro, MBLI-87 was as potent as GF120918 against ABCG2-mediated irinotecan efflux, and additionally was specific for ABCG2. Irinotecan 67-77 ATP binding cassette subfamily G member 2 (Junior blood group) Mus musculus 52-57 21205068-4 2011 The combination of low-dose irinotecan and US irradiation significantly inhibited the tube formation of HUVEC and vascular endothelial growth factor expression of tumor cells in vitro. Irinotecan 28-38 vascular endothelial growth factor A Homo sapiens 114-148 21617725-0 2011 Concurrence of UGT1A polymorphism and end-stage renal disease leads to severe toxicities of irinotecan in a patient with metastatic colon cancer. Irinotecan 92-102 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 15-20 21617725-9 2011 Sequencing analysis of the UGT1A genes found that the patient had variant alleles of UGT1A1*28, UGT1A1*60 and UGT1A9*22, which may lead to decreased glucuronidation and excretion of SN-38, and may account for increased irinotecan-related toxicity. Irinotecan 182-187 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 27-32 21617725-9 2011 Sequencing analysis of the UGT1A genes found that the patient had variant alleles of UGT1A1*28, UGT1A1*60 and UGT1A9*22, which may lead to decreased glucuronidation and excretion of SN-38, and may account for increased irinotecan-related toxicity. Irinotecan 182-187 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 85-91 21617725-9 2011 Sequencing analysis of the UGT1A genes found that the patient had variant alleles of UGT1A1*28, UGT1A1*60 and UGT1A9*22, which may lead to decreased glucuronidation and excretion of SN-38, and may account for increased irinotecan-related toxicity. Irinotecan 182-187 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 96-102 21617725-9 2011 Sequencing analysis of the UGT1A genes found that the patient had variant alleles of UGT1A1*28, UGT1A1*60 and UGT1A9*22, which may lead to decreased glucuronidation and excretion of SN-38, and may account for increased irinotecan-related toxicity. Irinotecan 182-187 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 110-116 21617725-9 2011 Sequencing analysis of the UGT1A genes found that the patient had variant alleles of UGT1A1*28, UGT1A1*60 and UGT1A9*22, which may lead to decreased glucuronidation and excretion of SN-38, and may account for increased irinotecan-related toxicity. Irinotecan 219-229 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 27-32 21617725-9 2011 Sequencing analysis of the UGT1A genes found that the patient had variant alleles of UGT1A1*28, UGT1A1*60 and UGT1A9*22, which may lead to decreased glucuronidation and excretion of SN-38, and may account for increased irinotecan-related toxicity. Irinotecan 219-229 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 85-91 21617725-9 2011 Sequencing analysis of the UGT1A genes found that the patient had variant alleles of UGT1A1*28, UGT1A1*60 and UGT1A9*22, which may lead to decreased glucuronidation and excretion of SN-38, and may account for increased irinotecan-related toxicity. Irinotecan 219-229 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 96-102 21617725-9 2011 Sequencing analysis of the UGT1A genes found that the patient had variant alleles of UGT1A1*28, UGT1A1*60 and UGT1A9*22, which may lead to decreased glucuronidation and excretion of SN-38, and may account for increased irinotecan-related toxicity. Irinotecan 219-229 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 110-116 21617725-11 2011 CONCLUSIONS: UGT1A polymorphisms and renal failure may lead to accumulation of SN-38, which may have played a role in the death of this patient. Irinotecan 79-84 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 13-18 21617725-12 2011 Irinotecan should be used cautiously in dialysis patients with metastatic colorectal cancer and screening for UGT1A polymorphisms may help in identifying patients with lower SN-38 glucuronidation rates and greater susceptibility to irinotecan-induced toxicity. Irinotecan 174-179 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 110-115 21617725-12 2011 Irinotecan should be used cautiously in dialysis patients with metastatic colorectal cancer and screening for UGT1A polymorphisms may help in identifying patients with lower SN-38 glucuronidation rates and greater susceptibility to irinotecan-induced toxicity. Irinotecan 232-242 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 110-115 21390185-8 2011 Axitinib and SN-38 combined treatment greatly inhibited the expression of the ATP7A and ABCG2 genes in endothelial and cancer cells, increasing the SN-38 intracellular concentration. Irinotecan 13-18 ATPase copper transporting alpha Homo sapiens 78-83 21390185-8 2011 Axitinib and SN-38 combined treatment greatly inhibited the expression of the ATP7A and ABCG2 genes in endothelial and cancer cells, increasing the SN-38 intracellular concentration. Irinotecan 13-18 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 88-93 21390185-8 2011 Axitinib and SN-38 combined treatment greatly inhibited the expression of the ATP7A and ABCG2 genes in endothelial and cancer cells, increasing the SN-38 intracellular concentration. Irinotecan 148-153 ATPase copper transporting alpha Homo sapiens 78-83 21390185-8 2011 Axitinib and SN-38 combined treatment greatly inhibited the expression of the ATP7A and ABCG2 genes in endothelial and cancer cells, increasing the SN-38 intracellular concentration. Irinotecan 148-153 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 88-93 21030469-11 2011 In addition, 7-ethyl-10-hydroxycamptothecin, a substrate of UGT1A1 (reported K(m) = 24 muM) seemed to be a weak inhibitor of bilirubin glucuronidation (IC(50) = 356.4 muM) but a partial inhibitor of estradiol-3-glucuronidation. Irinotecan 13-43 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 60-66 21030469-11 2011 In addition, 7-ethyl-10-hydroxycamptothecin, a substrate of UGT1A1 (reported K(m) = 24 muM) seemed to be a weak inhibitor of bilirubin glucuronidation (IC(50) = 356.4 muM) but a partial inhibitor of estradiol-3-glucuronidation. Irinotecan 13-43 latexin Homo sapiens 87-90 21030469-11 2011 In addition, 7-ethyl-10-hydroxycamptothecin, a substrate of UGT1A1 (reported K(m) = 24 muM) seemed to be a weak inhibitor of bilirubin glucuronidation (IC(50) = 356.4 muM) but a partial inhibitor of estradiol-3-glucuronidation. Irinotecan 13-43 latexin Homo sapiens 167-170 21194879-5 2011 The chemosensitivity of cell lines with various expression levels of PTEN was evaluated using 5-flurouracil (5-FU), oxaliplatin and irinotecan (CPT), and clinical significance was evaluated by immunohistochemical analysis of 133 CRC specimens. Irinotecan 132-142 phosphatase and tensin homolog Homo sapiens 69-73 21188390-0 2011 Predictive value of VEGF gene polymorphisms for metastatic colorectal cancer patients receiving first-line treatment including fluorouracil, irinotecan, and bevacizumab. Irinotecan 141-151 vascular endothelial growth factor A Homo sapiens 20-24 20177420-2 2011 Irinotecan"s active metabolite is inactivated by UDP-glucuronosyltransferase 1A1 (UGT1A1), which is deficient in Gilbert"s syndrome. Irinotecan 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 49-80 21159664-0 2011 Targeting the p38 MAPK pathway inhibits irinotecan resistance in colon adenocarcinoma. Irinotecan 40-50 mitogen-activated protein kinase 14 Homo sapiens 14-17 21159664-6 2011 Moreover, p38 inhibition sensitized tumor cells derived from both SN38-sensitive and -resistant HCT116 cells to irinotecan treatment in xenograft models. Irinotecan 112-122 mitogen-activated protein kinase 14 Homo sapiens 10-13 21159664-7 2011 Finally, we detected less phosphorylated p38 in primary colon cancer of patients sensitive to irinotecan-based treatment, compared with nonresponder patients. Irinotecan 94-104 mitogen-activated protein kinase 14 Homo sapiens 41-44 21159664-8 2011 This indicates that enhanced level of phosphorylated p38 could predict the absence of clinical response to irinotecan. Irinotecan 107-117 mitogen-activated protein kinase 14 Homo sapiens 53-56 21159664-9 2011 Altogether, our results show that the p38 MAPK pathway is involved in irinotecan sensitivity and suggest that phosphorylated p38 expression level could be used as a marker of clinical resistance to irinotecan. Irinotecan 70-80 mitogen-activated protein kinase 14 Homo sapiens 38-41 21159664-9 2011 Altogether, our results show that the p38 MAPK pathway is involved in irinotecan sensitivity and suggest that phosphorylated p38 expression level could be used as a marker of clinical resistance to irinotecan. Irinotecan 198-208 mitogen-activated protein kinase 14 Homo sapiens 38-41 21159664-9 2011 Altogether, our results show that the p38 MAPK pathway is involved in irinotecan sensitivity and suggest that phosphorylated p38 expression level could be used as a marker of clinical resistance to irinotecan. Irinotecan 198-208 mitogen-activated protein kinase 14 Homo sapiens 125-128 21159664-10 2011 They further suggest that targeting the p38 pathway may be a potential strategy to overcome resistance to irinotecan-based chemotherapies in colorectal cancer. Irinotecan 106-116 mitogen-activated protein kinase 14 Homo sapiens 40-43 20177420-2 2011 Irinotecan"s active metabolite is inactivated by UDP-glucuronosyltransferase 1A1 (UGT1A1), which is deficient in Gilbert"s syndrome. Irinotecan 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 82-88 20177420-3 2011 Irinotecan and metabolites are transported by P-glycoprotein, encoded by ABCB1. Irinotecan 0-10 ATP binding cassette subfamily B member 1 Homo sapiens 46-60 20177420-3 2011 Irinotecan and metabolites are transported by P-glycoprotein, encoded by ABCB1. Irinotecan 0-10 ATP binding cassette subfamily B member 1 Homo sapiens 73-78 21297369-4 2011 Irinotecan, now used for treatments of many cancers, is metabolically activated to SN-38 and then inactivated to SN-38 glucuronide by a UDP-glucuronosyltransferase UGT1A1. Irinotecan 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 164-170 20854796-9 2011 Cells incubated with anticancer drugs susceptible to glucuronidation, such as tamoxifen or irinotecan, together with methotrexate, showed a lesser degree of cytotoxicity, due to UGT1A6 induction. Irinotecan 91-101 UDP glucuronosyltransferase family 1 member A6 Homo sapiens 178-184 21283624-8 2011 We report that the phenotypes arising in mice globally deleted for the Cdc25s are due to the failure of small intestinal stem and progenitor cells to proliferate and that blocking cell division by inhibiting the cell cycle engine (through Cdc25 loss) versus by inducing DNA damage (via irinotecan) provokes a markedly different response of small intestinal epithelial cells. Irinotecan 286-296 cell division cycle 25C Mus musculus 71-76 20833148-5 2011 These studies confirm that hiCE demonstrates the most efficient kinetic parameters for CPT-11 activation, however, due to the high levels of hCE1 that are expressed in liver, the latter enzyme can contribute up to 50% of the total of drug hydrolysis in this tissue. Irinotecan 87-93 carboxylesterase 1 Homo sapiens 141-145 20833148-6 2011 Conversely, in human duodenum, jejunum, ileum and kidney, where hCE1 expression is very low, greater than 99% of the conversion of CPT-11 to SN-38 was mediated by hiCE. Irinotecan 141-146 carboxylesterase 1 Homo sapiens 64-68 20833148-8 2011 Overall, our studies demonstrate that hCE1 plays a significant role in CPT-11 hydrolysis even though it is up to 100-fold less efficient at drug activation than hiCE, and that drug activation in the intestine and kidney are likely major contributors to SN-38 production in vivo. Irinotecan 71-77 carboxylesterase 1 Homo sapiens 38-42 20680278-8 2011 CONCLUSIONS: Additional in vitro experiments suggest that these results can be explained by induction of UGT1A1 by methimazole, leading to higher SN-38G concentrations. Irinotecan 146-151 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 105-111 20833148-8 2011 Overall, our studies demonstrate that hCE1 plays a significant role in CPT-11 hydrolysis even though it is up to 100-fold less efficient at drug activation than hiCE, and that drug activation in the intestine and kidney are likely major contributors to SN-38 production in vivo. Irinotecan 253-258 carboxylesterase 1 Homo sapiens 38-42 20814789-0 2011 The small-molecule tyrosine kinase inhibitor nilotinib is a potent noncompetitive inhibitor of the SN-38 glucuronidation by human UGT1A1. Irinotecan 99-104 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 130-136 20354702-2 2011 CPT-11 is mainly activated to SN-38 by carboxylesterase (CES) and then detoxified to SN-38 glucuronide (SN-38G) by UDP-glucuronosyltransferase (UGT) in the liver. Irinotecan 0-6 UDP glycosyltransferase 2 family, polypeptide B Rattus norvegicus 115-142 20354702-2 2011 CPT-11 is mainly activated to SN-38 by carboxylesterase (CES) and then detoxified to SN-38 glucuronide (SN-38G) by UDP-glucuronosyltransferase (UGT) in the liver. Irinotecan 0-6 UDP glycosyltransferase 2 family, polypeptide B Rattus norvegicus 144-147 20354702-2 2011 CPT-11 is mainly activated to SN-38 by carboxylesterase (CES) and then detoxified to SN-38 glucuronide (SN-38G) by UDP-glucuronosyltransferase (UGT) in the liver. Irinotecan 85-90 UDP glycosyltransferase 2 family, polypeptide B Rattus norvegicus 115-142 20354702-2 2011 CPT-11 is mainly activated to SN-38 by carboxylesterase (CES) and then detoxified to SN-38 glucuronide (SN-38G) by UDP-glucuronosyltransferase (UGT) in the liver. Irinotecan 85-90 UDP glycosyltransferase 2 family, polypeptide B Rattus norvegicus 144-147 20354702-3 2011 SN-38G is excreted via bile and de-conjugated to SN-38 by beta-glucuronidase (beta-GLU) in the intestinal content. Irinotecan 0-5 glucuronidase, beta Rattus norvegicus 58-76 20354702-3 2011 SN-38G is excreted via bile and de-conjugated to SN-38 by beta-glucuronidase (beta-GLU) in the intestinal content. Irinotecan 0-5 glucuronidase, beta Rattus norvegicus 78-86 20814789-1 2011 PURPOSE: Inhibition of the UDP-glucuronosyltransferase (UGT) 1A1 by nilotinib was examined in vitro with SN-38 as a substrate, to estimate the possibility of drug-drug interaction of nilotinib with other medicines predominantly detoxified by UGT1A1. Irinotecan 105-110 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 27-64 20354702-3 2011 SN-38G is excreted via bile and de-conjugated to SN-38 by beta-glucuronidase (beta-GLU) in the intestinal content. Irinotecan 49-54 glucuronidase, beta Rattus norvegicus 58-76 20814789-2 2011 METHODS: Inhibition of UGT1A1-catalyzed SN-38 glucuronidation by nilotinib was examined with human liver microsomes (HLM) and recombinant human UGT1A1 as enzyme sources. Irinotecan 40-45 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 23-29 20354702-3 2011 SN-38G is excreted via bile and de-conjugated to SN-38 by beta-glucuronidase (beta-GLU) in the intestinal content. Irinotecan 49-54 glucuronidase, beta Rattus norvegicus 78-86 20814789-4 2011 RESULTS: Nilotinib potently inhibited the SN-38 glucuronidation by human liver microsomal UGT1A1 and recombinant UGT1A1 in a noncompetitive manner, with K(i) values of 0.286 +- 0.0094 and 0.079 +- 0.0029 muM, respectively. Irinotecan 42-47 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 90-96 20814789-4 2011 RESULTS: Nilotinib potently inhibited the SN-38 glucuronidation by human liver microsomal UGT1A1 and recombinant UGT1A1 in a noncompetitive manner, with K(i) values of 0.286 +- 0.0094 and 0.079 +- 0.0029 muM, respectively. Irinotecan 42-47 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 113-119 21609311-3 2011 SN-38 collapsed the mitochondrial membrane potential (MMP), arrested cells in S- and G2-phases of the cell cycle, and induced apoptosis via activation of caspase 3 and PARP. Irinotecan 0-5 caspase 3 Homo sapiens 154-163 21634054-0 2011 Efficacy of irinotecan in combination with 5-fluorouracil (FOLFIRI) for metastatic gastric or gastroesophageal junction adenocarcinomas (MGA) treatment. Irinotecan 12-22 MAX dimerization protein MGA Homo sapiens 139-142 22149660-8 2011 RESULTS: We found a significant inhibition of MATE1-mediated MPP uptake by several antineoplastic agents and pH dependent IC(50)values for mitoxantrone (7.8 muM at pH 7.4 and 0.6 muM at pH 8.5) as well as for irinotecan (4.4 muM at pH 7.4 and 1.1 muM at pH 8.5), respectively. Irinotecan 209-219 solute carrier family 47 member 1 Homo sapiens 46-51 21282945-10 2011 SDHA and GAPDH were the most variable genes in the jejunum and colon where they were 4.4 and 3.2 fold upregulated following irinotecan, respectively. Irinotecan 124-134 succinate dehydrogenase complex flavoprotein subunit A Rattus norvegicus 0-4 21282945-10 2011 SDHA and GAPDH were the most variable genes in the jejunum and colon where they were 4.4 and 3.2 fold upregulated following irinotecan, respectively. Irinotecan 124-134 glyceraldehyde-3-phosphate dehydrogenase Rattus norvegicus 9-14 21609311-3 2011 SN-38 collapsed the mitochondrial membrane potential (MMP), arrested cells in S- and G2-phases of the cell cycle, and induced apoptosis via activation of caspase 3 and PARP. Irinotecan 0-5 poly(ADP-ribose) polymerase 1 Homo sapiens 168-172 21212430-0 2011 The role of HER-3 expression in the prediction of clinical outcome for advanced colorectal cancer patients receiving irinotecan and cetuximab. Irinotecan 117-127 erb-b2 receptor tyrosine kinase 3 Homo sapiens 12-17 22016648-9 2011 The acute heart and kidney toxicity of the active metabolite SN-38 from its less toxic prodrug, irinotecan could be differentiated, and two novel gene markers for hormone replacement therapy were identified, namely fabp4 and pparg, which were down-regulated by estradiol treatment. Irinotecan 61-66 fatty acid binding protein 4, adipocyte Mus musculus 215-220 22016648-9 2011 The acute heart and kidney toxicity of the active metabolite SN-38 from its less toxic prodrug, irinotecan could be differentiated, and two novel gene markers for hormone replacement therapy were identified, namely fabp4 and pparg, which were down-regulated by estradiol treatment. Irinotecan 61-66 peroxisome proliferator activated receptor gamma Mus musculus 225-230 21733405-0 2011 In Situ intestinal perfusion of irinotecan: application to P-gp mediated drug interaction and introduction of an improved HPLC assay. Irinotecan 32-42 phosphoglycolate phosphatase Rattus norvegicus 59-63 21733405-1 2011 PURPOSE: To determine experimentally the intestinal permeability of the anticancer prodrug irinotecan, and to quantify the amount of its cytotoxic metabolite SN-38 that is intestinally excreted (exsorped) as a predictor of intestinal toxicity, and to assess the effect of p-glycoprotein (p-gp) inhibitors (verapamil as a model) on the permeability and toxicity of irinotecan. Irinotecan 158-163 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 272-286 21733405-1 2011 PURPOSE: To determine experimentally the intestinal permeability of the anticancer prodrug irinotecan, and to quantify the amount of its cytotoxic metabolite SN-38 that is intestinally excreted (exsorped) as a predictor of intestinal toxicity, and to assess the effect of p-glycoprotein (p-gp) inhibitors (verapamil as a model) on the permeability and toxicity of irinotecan. Irinotecan 158-163 phosphoglycolate phosphatase Rattus norvegicus 288-292 21733405-9 2011 P-gp inhibition using verapamil found to significantly enhance the intestinal permeability of irinotecan and potentially decrease the intestinal toxicity due to SN-38 exposure. Irinotecan 94-104 phosphoglycolate phosphatase Rattus norvegicus 0-4 21733405-9 2011 P-gp inhibition using verapamil found to significantly enhance the intestinal permeability of irinotecan and potentially decrease the intestinal toxicity due to SN-38 exposure. Irinotecan 161-166 phosphoglycolate phosphatase Rattus norvegicus 0-4 21150467-13 2011 The UGT1A1*6 and UGT1A1*27 genotypes might be useful predictors of grade 4 neutropenia in patients who receive irinotecan-based chemotherapy. Irinotecan 111-121 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 21150467-13 2011 The UGT1A1*6 and UGT1A1*27 genotypes might be useful predictors of grade 4 neutropenia in patients who receive irinotecan-based chemotherapy. Irinotecan 111-121 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 17-23 21473286-2 2011 Especially, SNPs of UTG1A1 and CYP2C19 are important for patients who are treated with irinotecan and proton pump inhibitors, respectively. Irinotecan 87-97 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 31-38 21212430-3 2011 The aim of our analysis was to investigate a possible correlation between HER-3 expression and clinical outcome in wild-type K-RAS advanced colorectal cancer patients receiving cetuximab and irinotecan. Irinotecan 191-201 erb-b2 receptor tyrosine kinase 3 Homo sapiens 74-79 21212430-3 2011 The aim of our analysis was to investigate a possible correlation between HER-3 expression and clinical outcome in wild-type K-RAS advanced colorectal cancer patients receiving cetuximab and irinotecan. Irinotecan 191-201 KRAS proto-oncogene, GTPase Homo sapiens 125-130 21985855-0 2011 Successful treatment of a patient with HER2-positive metastatic gastric cancer with third-line combination therapy with irinotecan, 5-fluorouracil, leucovorin and trastuzumab (FOLFIRI-T). Irinotecan 120-130 erb-b2 receptor tyrosine kinase 2 Homo sapiens 39-43 21212430-4 2011 We retrospectively analyzed immunoreactivity for HER-3 in wild-type K-RAS advanced colorectal cancer patients receiving irinotecan and cetuximab. Irinotecan 120-130 erb-b2 receptor tyrosine kinase 3 Homo sapiens 49-54 20872237-8 2010 We found that a clinically approved camptothecin analog, irinotecan suppressed the migration, Cdc42 activity, and autophosphorylation of FAK, and attenuated integrin beta1 distribution selectively in LM8 cells. Irinotecan 57-67 cell division cycle 42 Mus musculus 94-99 21674030-0 2011 Strain- and sex-dependent circadian changes in abcc2 transporter expression: implications for irinotecan chronotolerance in mouse ileum. Irinotecan 94-104 ATP-binding cassette, sub-family C (CFTR/MRP), member 2 Mus musculus 47-52 21674030-1 2011 BACKGROUND: ATP-binding cassette transporter abcc2 is involved in the cellular efflux of irinotecan. Irinotecan 89-99 ATP-binding cassette, sub-family C (CFTR/MRP), member 2 Mus musculus 45-50 21674030-3 2011 Here, we investigate whether circadian changes in local abcc2 expression participate in the circadian rhythm of irinotecan toxicity for ileum mucosa, and further assess whether genetic background or sex modify this relation. Irinotecan 112-122 ATP-binding cassette, sub-family C (CFTR/MRP), member 2 Mus musculus 56-61 21674030-13 2011 CONCLUSIONS/SIGNIFICANCE: Strain- and sex-dependent circadian patterns in abcc2 expressions displayed robust relations with the chronotolerance of ileum mucosa for irinotecan. Irinotecan 164-174 ATP-binding cassette, sub-family C (CFTR/MRP), member 2 Mus musculus 74-79 21355292-9 2011 CONCLUSION: GSTA and ZNF subunit genes might play an important regulation role in the irinotecan resistance of colon cancer. Irinotecan 86-96 zinc finger protein 763 Homo sapiens 21-24 22041697-3 2011 Organic anion transporting polypeptide 1B1 (OATP1B1, gene SLCO1B1) is expressed on the basolateral membrane of hepatocytes and can facilitate hepatic uptake of certain clinically relevant drugs such as statins except for fluvastatin, angiotensin converting enzyme inhibitors, angiotensin II receptor antagonists, antidiabetic drug (repaglinide) and anticancer drugs (SN-38 and methotrexate). Irinotecan 367-372 solute carrier organic anion transporter family member 1B1 Homo sapiens 0-42 22041697-3 2011 Organic anion transporting polypeptide 1B1 (OATP1B1, gene SLCO1B1) is expressed on the basolateral membrane of hepatocytes and can facilitate hepatic uptake of certain clinically relevant drugs such as statins except for fluvastatin, angiotensin converting enzyme inhibitors, angiotensin II receptor antagonists, antidiabetic drug (repaglinide) and anticancer drugs (SN-38 and methotrexate). Irinotecan 367-372 solute carrier organic anion transporter family member 1B1 Homo sapiens 44-51 22041697-3 2011 Organic anion transporting polypeptide 1B1 (OATP1B1, gene SLCO1B1) is expressed on the basolateral membrane of hepatocytes and can facilitate hepatic uptake of certain clinically relevant drugs such as statins except for fluvastatin, angiotensin converting enzyme inhibitors, angiotensin II receptor antagonists, antidiabetic drug (repaglinide) and anticancer drugs (SN-38 and methotrexate). Irinotecan 367-372 solute carrier organic anion transporter family member 1B1 Homo sapiens 58-65 22041697-5 2011 For examples, the SLCO1B1*15 haplotype (or 521T>C genotype) results in decreased uptake activity of SN-38 from systemic circulation, leading to increased plasma concentration of SN-38 and an enhanced risk of neutropenia. Irinotecan 103-108 solute carrier organic anion transporter family member 1B1 Homo sapiens 18-25 22041697-5 2011 For examples, the SLCO1B1*15 haplotype (or 521T>C genotype) results in decreased uptake activity of SN-38 from systemic circulation, leading to increased plasma concentration of SN-38 and an enhanced risk of neutropenia. Irinotecan 181-186 solute carrier organic anion transporter family member 1B1 Homo sapiens 18-25 21208842-1 2010 BACKGROUND: The aim of this multicenter phase II study was to demonstrate the activity of the epidermal growth factor receptor (EGFR)-targeting monoclonal antibody cetuximab combined with irinotecan in the treatment of Latin American patients with EGFR-expressing metastatic colorectal cancer (mCRC) in whom previous treatment with an irinotecan-containing regimen had failed. Irinotecan 335-345 epidermal growth factor receptor Homo sapiens 94-126 21208842-1 2010 BACKGROUND: The aim of this multicenter phase II study was to demonstrate the activity of the epidermal growth factor receptor (EGFR)-targeting monoclonal antibody cetuximab combined with irinotecan in the treatment of Latin American patients with EGFR-expressing metastatic colorectal cancer (mCRC) in whom previous treatment with an irinotecan-containing regimen had failed. Irinotecan 335-345 epidermal growth factor receptor Homo sapiens 128-132 20872237-8 2010 We found that a clinically approved camptothecin analog, irinotecan suppressed the migration, Cdc42 activity, and autophosphorylation of FAK, and attenuated integrin beta1 distribution selectively in LM8 cells. Irinotecan 57-67 PTK2 protein tyrosine kinase 2 Mus musculus 137-140 20872237-8 2010 We found that a clinically approved camptothecin analog, irinotecan suppressed the migration, Cdc42 activity, and autophosphorylation of FAK, and attenuated integrin beta1 distribution selectively in LM8 cells. Irinotecan 57-67 integrin beta 1 (fibronectin receptor beta) Mus musculus 157-171 20847137-3 2010 In this study, catalytic activities of the two alleles found in East Asians, CYP3A4*16 (T185S) and CYP3A4*18 (L293P), were assessed using the following seven substrates: midazolam, carbamazepine, atorvastatin, paclitaxel, docetaxel, irinotecan, and terfenadine. Irinotecan 233-243 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 77-83 20583968-1 2010 The ATP-binding cassette transporters P-glycoprotein (ABCB1, MDR1) and multidrug resistance protein 4 (MRP4) efflux irinotecan and its active metabolite SN-38 in vitro, and thus may contribute to system clearance of these compounds. Irinotecan 116-126 ATP-binding cassette, sub-family B (MDR/TAP), member 1B Mus musculus 61-65 20583968-1 2010 The ATP-binding cassette transporters P-glycoprotein (ABCB1, MDR1) and multidrug resistance protein 4 (MRP4) efflux irinotecan and its active metabolite SN-38 in vitro, and thus may contribute to system clearance of these compounds. Irinotecan 116-126 ATP-binding cassette, sub-family C (CFTR/MRP), member 4 Mus musculus 71-101 20583968-1 2010 The ATP-binding cassette transporters P-glycoprotein (ABCB1, MDR1) and multidrug resistance protein 4 (MRP4) efflux irinotecan and its active metabolite SN-38 in vitro, and thus may contribute to system clearance of these compounds. Irinotecan 116-126 ATP-binding cassette, sub-family C (CFTR/MRP), member 4 Mus musculus 103-107 20583968-1 2010 The ATP-binding cassette transporters P-glycoprotein (ABCB1, MDR1) and multidrug resistance protein 4 (MRP4) efflux irinotecan and its active metabolite SN-38 in vitro, and thus may contribute to system clearance of these compounds. Irinotecan 153-158 ATP-binding cassette, sub-family B (MDR/TAP), member 1B Mus musculus 54-59 20583968-1 2010 The ATP-binding cassette transporters P-glycoprotein (ABCB1, MDR1) and multidrug resistance protein 4 (MRP4) efflux irinotecan and its active metabolite SN-38 in vitro, and thus may contribute to system clearance of these compounds. Irinotecan 116-126 ATP-binding cassette, sub-family B (MDR/TAP), member 1B Mus musculus 54-59 20847137-3 2010 In this study, catalytic activities of the two alleles found in East Asians, CYP3A4*16 (T185S) and CYP3A4*18 (L293P), were assessed using the following seven substrates: midazolam, carbamazepine, atorvastatin, paclitaxel, docetaxel, irinotecan, and terfenadine. Irinotecan 233-243 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 99-105 20583968-1 2010 The ATP-binding cassette transporters P-glycoprotein (ABCB1, MDR1) and multidrug resistance protein 4 (MRP4) efflux irinotecan and its active metabolite SN-38 in vitro, and thus may contribute to system clearance of these compounds. Irinotecan 153-158 ATP-binding cassette, sub-family B (MDR/TAP), member 1B Mus musculus 61-65 20583968-1 2010 The ATP-binding cassette transporters P-glycoprotein (ABCB1, MDR1) and multidrug resistance protein 4 (MRP4) efflux irinotecan and its active metabolite SN-38 in vitro, and thus may contribute to system clearance of these compounds. Irinotecan 153-158 ATP-binding cassette, sub-family C (CFTR/MRP), member 4 Mus musculus 71-101 20847137-5 2010 CYP3A4.16 exhibited intrinsic clearances (V(max)/K(m)) that were lowered considerably (by 84-60%) for metabolism of midazolam, carbamazepine, atorvastatin, paclitaxel, and irinotecan compared with CYP3A4.1 due to increased K(m) with or without decreased V(max) values, whereas no apparent decrease in intrinsic clearance was observed for docetaxel. Irinotecan 172-182 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 0-6 20583968-1 2010 The ATP-binding cassette transporters P-glycoprotein (ABCB1, MDR1) and multidrug resistance protein 4 (MRP4) efflux irinotecan and its active metabolite SN-38 in vitro, and thus may contribute to system clearance of these compounds. Irinotecan 153-158 ATP-binding cassette, sub-family C (CFTR/MRP), member 4 Mus musculus 103-107 20583968-4 2010 Mdr1a/b genotype was a significant covariate for the clearance of both irinotecan lactone and SN-38 lactone. Irinotecan 71-89 ATP-binding cassette, sub-family B (MDR/TAP), member 1A Mus musculus 0-5 20847137-5 2010 CYP3A4.16 exhibited intrinsic clearances (V(max)/K(m)) that were lowered considerably (by 84-60%) for metabolism of midazolam, carbamazepine, atorvastatin, paclitaxel, and irinotecan compared with CYP3A4.1 due to increased K(m) with or without decreased V(max) values, whereas no apparent decrease in intrinsic clearance was observed for docetaxel. Irinotecan 172-182 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 197-203 20583968-4 2010 Mdr1a/b genotype was a significant covariate for the clearance of both irinotecan lactone and SN-38 lactone. Irinotecan 94-107 ATP-binding cassette, sub-family B (MDR/TAP), member 1A Mus musculus 0-5 20583968-5 2010 Exposures to irinotecan lactone and SN-38 lactone after a 40 mg/kg dose were 1.6-fold higher in Mdr1a/b(-/-) mice compared to wild-type mice. Irinotecan 13-31 ATP-binding cassette, sub-family B (MDR/TAP), member 1A Mus musculus 96-101 20583968-5 2010 Exposures to irinotecan lactone and SN-38 lactone after a 40 mg/kg dose were 1.6-fold higher in Mdr1a/b(-/-) mice compared to wild-type mice. Irinotecan 36-49 ATP-binding cassette, sub-family B (MDR/TAP), member 1A Mus musculus 96-101 20583968-7 2010 These results suggest that P-gp plays a role in irinotecan and SN-38 elimination, but Mrp4 does not affect irinotecan or SN-38 plasma pharmacokinetics. Irinotecan 48-58 phosphoglycolate phosphatase Mus musculus 27-31 20583968-7 2010 These results suggest that P-gp plays a role in irinotecan and SN-38 elimination, but Mrp4 does not affect irinotecan or SN-38 plasma pharmacokinetics. Irinotecan 63-68 phosphoglycolate phosphatase Mus musculus 27-31 20847137-6 2010 On the other hand, K(m) values for CYP3A4.18 were comparable to those for CYP3A4.1 for all substrates except terfenadine; but V(max) values were lower for midazolam, paclitaxel, docetaxel, and irinotecan, resulting in partially reduced intrinsic clearance values (by 34-52%). Irinotecan 193-203 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 35-41 20889729-8 2010 This synergy was not universal but, when observed (Susa S/R, H1975, H358, and MDA-MB-231 cell lines), was related to SN-38 intracellular accumulation (2.2- to 4.8-fold increase, P < 0.05 for each), attributable to the inhibition of the breast cancer-related protein (BCRP) efflux pump by lapatinib. Irinotecan 117-122 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 239-268 20198621-0 2010 Synergistic antitumor activity of the SN-38-incorporating polymeric micelles NK012 with S-1 in a mouse model of non-small cell lung cancer. Irinotecan 38-43 proteasome (prosome, macropain) 26S subunit, non-ATPase, 1 Mus musculus 88-91 20198621-1 2010 The combination therapy of CPT-11, a prodrug of SN-38, with S-1, a dihydropyrimidine dehydrogenase inhibitory fluoropyrimidine, shows a high clinical response rate in non-small cell lung cancer (NSCLC). Irinotecan 27-33 proteasome (prosome, macropain) 26S subunit, non-ATPase, 1 Mus musculus 60-63 19862647-0 2010 MDR1 polymorphism role in patients treated with cetuximab and irinotecan in irinotecan refractory colorectal cancer. Irinotecan 62-72 ATP binding cassette subfamily B member 1 Homo sapiens 0-4 19862647-0 2010 MDR1 polymorphism role in patients treated with cetuximab and irinotecan in irinotecan refractory colorectal cancer. Irinotecan 76-86 ATP binding cassette subfamily B member 1 Homo sapiens 0-4 19862647-1 2010 The aim of the study was to evaluate the influence of the MDR1 C3435T polymorphism on the therapeutic response in 23 patients treated with cetuximab plus irinotecan for irinotecan refractory liver metastatic colorectal cancer considering their KRAS status. Irinotecan 154-164 ATP binding cassette subfamily B member 1 Homo sapiens 58-62 19862647-1 2010 The aim of the study was to evaluate the influence of the MDR1 C3435T polymorphism on the therapeutic response in 23 patients treated with cetuximab plus irinotecan for irinotecan refractory liver metastatic colorectal cancer considering their KRAS status. Irinotecan 169-179 ATP binding cassette subfamily B member 1 Homo sapiens 58-62 19862647-2 2010 Indeed, irinotecan and its active metabolite (SN-38) are both substrates of P-glycoprotein (P-gp) encoded by MDR1. Irinotecan 8-18 ATP binding cassette subfamily B member 1 Homo sapiens 76-90 19862647-2 2010 Indeed, irinotecan and its active metabolite (SN-38) are both substrates of P-glycoprotein (P-gp) encoded by MDR1. Irinotecan 8-18 ATP binding cassette subfamily B member 1 Homo sapiens 92-96 19862647-2 2010 Indeed, irinotecan and its active metabolite (SN-38) are both substrates of P-glycoprotein (P-gp) encoded by MDR1. Irinotecan 8-18 ATP binding cassette subfamily B member 1 Homo sapiens 109-113 19862647-2 2010 Indeed, irinotecan and its active metabolite (SN-38) are both substrates of P-glycoprotein (P-gp) encoded by MDR1. Irinotecan 46-51 ATP binding cassette subfamily B member 1 Homo sapiens 76-90 19862647-2 2010 Indeed, irinotecan and its active metabolite (SN-38) are both substrates of P-glycoprotein (P-gp) encoded by MDR1. Irinotecan 46-51 ATP binding cassette subfamily B member 1 Homo sapiens 92-96 19862647-2 2010 Indeed, irinotecan and its active metabolite (SN-38) are both substrates of P-glycoprotein (P-gp) encoded by MDR1. Irinotecan 46-51 ATP binding cassette subfamily B member 1 Homo sapiens 109-113 19862647-9 2010 These results suggest that EGFR inhibition by cetuximab may overcome this irinotecan resistance by abrogating drug efflux depending on MDR1 3435 polymorphism. Irinotecan 74-84 epidermal growth factor receptor Homo sapiens 27-31 19862647-9 2010 These results suggest that EGFR inhibition by cetuximab may overcome this irinotecan resistance by abrogating drug efflux depending on MDR1 3435 polymorphism. Irinotecan 74-84 ATP binding cassette subfamily B member 1 Homo sapiens 135-139 20889729-8 2010 This synergy was not universal but, when observed (Susa S/R, H1975, H358, and MDA-MB-231 cell lines), was related to SN-38 intracellular accumulation (2.2- to 4.8-fold increase, P < 0.05 for each), attributable to the inhibition of the breast cancer-related protein (BCRP) efflux pump by lapatinib. Irinotecan 117-122 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 270-274 21151408-2 2010 This paper uses a case-based approach to summarize the consensus recommendations developed during that meeting.These are the consensus recommendations:Testing for the KRAS status of the tumour should be performed as soon as an egfr inhibitor is being considered as an option for treatment.Anti-egfr therapies are not recommended for the treatment of patients with tumours showing mutated KRAS status.For a patient with wild-type KRAS and an Eastern Cooperative Oncology Group status of 0-2, whose mcrc has previously been treated with a fluoropyrimidine, irinotecan, and oxaliplatin, switching to an egfr inhibitor is a recommended strategy.Cetuximab, cetuximab plus irinotecan, and panitumumab are all options for third-line therapy in patients with wild-type KRAS, provided that tolerability is acceptable. Irinotecan 555-565 KRAS proto-oncogene, GTPase Homo sapiens 167-171 21042746-5 2010 Combinations of CG2 with SN38 (the active form of irinotecan), 5FU, or oxaliplatin were more effective than the agents alone when used to inhibit the growth of HCT116 cells. Irinotecan 50-60 transmembrane protein 245 Homo sapiens 16-19 21042746-6 2010 The protein expressions of acetyl-H3, p21, caspase-3, -8, and -9, PARP, and XIAP were affected in a time- and dose-dependent manner in HCT116 cells treated with the CG2 alone or combined CG2 and SN-38. Irinotecan 195-200 caspase 3 Homo sapiens 43-64 21042746-6 2010 The protein expressions of acetyl-H3, p21, caspase-3, -8, and -9, PARP, and XIAP were affected in a time- and dose-dependent manner in HCT116 cells treated with the CG2 alone or combined CG2 and SN-38. Irinotecan 195-200 transmembrane protein 245 Homo sapiens 165-168 21151408-2 2010 This paper uses a case-based approach to summarize the consensus recommendations developed during that meeting.These are the consensus recommendations:Testing for the KRAS status of the tumour should be performed as soon as an egfr inhibitor is being considered as an option for treatment.Anti-egfr therapies are not recommended for the treatment of patients with tumours showing mutated KRAS status.For a patient with wild-type KRAS and an Eastern Cooperative Oncology Group status of 0-2, whose mcrc has previously been treated with a fluoropyrimidine, irinotecan, and oxaliplatin, switching to an egfr inhibitor is a recommended strategy.Cetuximab, cetuximab plus irinotecan, and panitumumab are all options for third-line therapy in patients with wild-type KRAS, provided that tolerability is acceptable. Irinotecan 667-677 KRAS proto-oncogene, GTPase Homo sapiens 167-171 21472350-1 2010 Irinotecan (CTP-11) is a topoisomerase I inhibitor used in the treatment of colorectal cancer and non-small cell lung cancer (NSCLC). Irinotecan 0-10 SPANX family member C Homo sapiens 12-18 20829196-7 2010 Interestingly, multidrug-resistant cell lines that overexpressed P-glycoprotein (P-gp), multidrug resistance-associated proteins, and breast cancer resistance protein were rendered resistant to docetaxel, vinblastine, SN-38, and doxorubicin, but not to I-387. Irinotecan 218-223 ATP binding cassette subfamily B member 1 Homo sapiens 65-79 20829196-7 2010 Interestingly, multidrug-resistant cell lines that overexpressed P-glycoprotein (P-gp), multidrug resistance-associated proteins, and breast cancer resistance protein were rendered resistant to docetaxel, vinblastine, SN-38, and doxorubicin, but not to I-387. Irinotecan 218-223 ATP binding cassette subfamily B member 1 Homo sapiens 81-85 20829196-7 2010 Interestingly, multidrug-resistant cell lines that overexpressed P-glycoprotein (P-gp), multidrug resistance-associated proteins, and breast cancer resistance protein were rendered resistant to docetaxel, vinblastine, SN-38, and doxorubicin, but not to I-387. Irinotecan 218-223 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 134-166 21472350-3 2010 We isolated irinotecan-resistant human NSCLC A549 cells, termed A549/CTP-11R cells. Irinotecan 12-22 SPANX family member C Homo sapiens 69-75 21472350-4 2010 A549/CTP-11R cells were resistant to irinotecan, as well as paclitaxel, gemcitabine and carboplatin. Irinotecan 37-47 SPANX family member C Homo sapiens 5-11 21472350-5 2010 Curcumin, a nuclear factor-kappaB (NF-kappaB) inhibitor, increased the sensitivity to irinotecan of A549/CTP-11R cells. Irinotecan 86-96 nuclear factor kappa B subunit 1 Homo sapiens 35-44 21472350-5 2010 Curcumin, a nuclear factor-kappaB (NF-kappaB) inhibitor, increased the sensitivity to irinotecan of A549/CTP-11R cells. Irinotecan 86-96 SPANX family member C Homo sapiens 105-111 20039040-1 2010 BACKGROUND: Expression of the DNA repair protein O (6)-methylguanine-DNA methyltransferase (MGMT) correlates with resistance to irinotecan in colorectal cancer cell lines. Irinotecan 128-138 O-6-methylguanine-DNA methyltransferase Homo sapiens 92-96 20924375-0 2010 Activity of irinotecan and temozolomide in the presence of O6-methylguanine-DNA methyltransferase inhibition in neuroblastoma pre-clinical models. Irinotecan 12-22 O-6-methylguanine-DNA methyltransferase Homo sapiens 59-97 20924375-3 2010 Using neuroblastoma pre-clinical models, we determined whether the inhibition of MGMT by O(6)-benzylguanine (O6-BG) could enhance the anti-tumour activity of TMZ and irinotecan. Irinotecan 166-176 O-6-methylguanine-DNA methyltransferase Homo sapiens 81-85 19967559-1 2010 Breast cancer resistance protein (BCRP/ABCG2), an ATP-binding cassette half transporter, confers multidrug resistance (MDR) to a series of antitumor agents such as mitoxantrone, daunorubicin, SN-38, and topotecan, and often limits the efficacy of chemotherapy. Irinotecan 192-197 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 0-32 19967559-1 2010 Breast cancer resistance protein (BCRP/ABCG2), an ATP-binding cassette half transporter, confers multidrug resistance (MDR) to a series of antitumor agents such as mitoxantrone, daunorubicin, SN-38, and topotecan, and often limits the efficacy of chemotherapy. Irinotecan 192-197 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 34-38 19967559-1 2010 Breast cancer resistance protein (BCRP/ABCG2), an ATP-binding cassette half transporter, confers multidrug resistance (MDR) to a series of antitumor agents such as mitoxantrone, daunorubicin, SN-38, and topotecan, and often limits the efficacy of chemotherapy. Irinotecan 192-197 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 39-44 20099280-0 2010 Insulin-like growth factor 1 expression correlates with clinical outcome in K-RAS wild type colorectal cancer patients treated with cetuximab and irinotecan. Irinotecan 146-156 insulin like growth factor 1 Homo sapiens 0-28 20099280-0 2010 Insulin-like growth factor 1 expression correlates with clinical outcome in K-RAS wild type colorectal cancer patients treated with cetuximab and irinotecan. Irinotecan 146-156 KRAS proto-oncogene, GTPase Homo sapiens 76-81 20099280-4 2010 IGF-1 expression and K-RAS mutational status was assessed in advanced colorectal cancer patients receiving irinotecan/cetuximab. Irinotecan 107-117 insulin like growth factor 1 Homo sapiens 0-5 20099280-9 2010 Among K-RAS wild type patients, IGF-1 negative and positive tumors showed a partial response to cetuximab-irinotecan in 13 (65%) and 11 (22%) cases, respectively (p = 0.002). Irinotecan 106-116 insulin like growth factor 1 Homo sapiens 32-37 21036743-4 2010 RESULTS: High intratumoral gene expression levels of EGFR, VEGFR2 and NRP1 were associated with longer overall survival (OS) in patients receiving combined monoclonal antibodies with or without irinotecan. Irinotecan 194-204 epidermal growth factor receptor Homo sapiens 53-57 21036743-4 2010 RESULTS: High intratumoral gene expression levels of EGFR, VEGFR2 and NRP1 were associated with longer overall survival (OS) in patients receiving combined monoclonal antibodies with or without irinotecan. Irinotecan 194-204 kinase insert domain receptor Homo sapiens 59-65 21036743-4 2010 RESULTS: High intratumoral gene expression levels of EGFR, VEGFR2 and NRP1 were associated with longer overall survival (OS) in patients receiving combined monoclonal antibodies with or without irinotecan. Irinotecan 194-204 neuropilin 1 Homo sapiens 70-74 20066420-0 2010 Se-methylselenocysteine sensitizes hypoxic tumor cells to irinotecan by targeting hypoxia-inducible factor 1alpha. Irinotecan 58-68 hypoxia inducible factor 1 subunit alpha Homo sapiens 82-113 20066420-8 2010 CONCLUSIONS: Our results show that HIF-1alpha is a critical target for MSC and its inhibition was associated with enhanced antitumor activity of irinotecan. Irinotecan 145-155 hypoxia inducible factor 1 subunit alpha Homo sapiens 35-45 20811661-0 2010 Irinotecan overcomes the resistance to 5-fluorouracil in human colon cancer xenografts by down-regulation of intratumoral thymidylate synthase. Irinotecan 0-10 thymidylate synthetase Homo sapiens 122-142 20682600-4 2010 This study aims to characterize the expression of multiple MMPs including MMP-1, -2, -3, -9 and -12 and their inhibitors, tissue inhibitor of metalloproteinase (TIMP)-1 and -2, in a rat model of irinotecan-induced mucositis. Irinotecan 195-205 matrix metallopeptidase 1 Rattus norvegicus 59-63 20682600-4 2010 This study aims to characterize the expression of multiple MMPs including MMP-1, -2, -3, -9 and -12 and their inhibitors, tissue inhibitor of metalloproteinase (TIMP)-1 and -2, in a rat model of irinotecan-induced mucositis. Irinotecan 195-205 matrix metallopeptidase 1 Rattus norvegicus 74-99 20682600-4 2010 This study aims to characterize the expression of multiple MMPs including MMP-1, -2, -3, -9 and -12 and their inhibitors, tissue inhibitor of metalloproteinase (TIMP)-1 and -2, in a rat model of irinotecan-induced mucositis. Irinotecan 195-205 TIMP metallopeptidase inhibitor 1 Rattus norvegicus 122-175 20682600-8 2010 A significant alteration in both gene expression and tissue levels of MMPs and TIMPs was noted following irinotecan. Irinotecan 105-115 matrix metallopeptidase 1 Rattus norvegicus 70-74 20682600-11 2010 TIMP-1 and -2 levels were significantly altered in the jejunum following irinotecan. Irinotecan 73-83 TIMP metallopeptidase inhibitor 1 Rattus norvegicus 0-13 20682600-12 2010 The augmentation in the expression profiles of MMPs and their inhibitors correlated with histopathological alterations observed in the tissue following irinotecan. Irinotecan 152-162 matrix metallopeptidase 1 Rattus norvegicus 47-51 20811661-6 2010 CPT-11, but not l-OHP, induced a decrease in activities and protein levels of TS and an increase in those of RNR in KM12C/5-FU tumors only, which was likely related to decreased expressions of several proteins in G1/S phase of the cells including CDK4, pRB, and E2F1 in these tumors. Irinotecan 0-6 thymidylate synthetase Homo sapiens 78-80 20811661-6 2010 CPT-11, but not l-OHP, induced a decrease in activities and protein levels of TS and an increase in those of RNR in KM12C/5-FU tumors only, which was likely related to decreased expressions of several proteins in G1/S phase of the cells including CDK4, pRB, and E2F1 in these tumors. Irinotecan 0-6 cyclin dependent kinase 4 Homo sapiens 247-251 20811661-6 2010 CPT-11, but not l-OHP, induced a decrease in activities and protein levels of TS and an increase in those of RNR in KM12C/5-FU tumors only, which was likely related to decreased expressions of several proteins in G1/S phase of the cells including CDK4, pRB, and E2F1 in these tumors. Irinotecan 0-6 RB transcriptional corepressor 1 Homo sapiens 253-256 20811661-6 2010 CPT-11, but not l-OHP, induced a decrease in activities and protein levels of TS and an increase in those of RNR in KM12C/5-FU tumors only, which was likely related to decreased expressions of several proteins in G1/S phase of the cells including CDK4, pRB, and E2F1 in these tumors. Irinotecan 0-6 E2F transcription factor 1 Homo sapiens 262-266 20716241-1 2010 WHAT IS ALREADY KNOWN ABOUT THIS SUBJECT: The anticancer agent irinotecan is a prodrug that is hydrolyzed by hepatic carboxylesterase to its active and toxic metabolite SN-38 and oxidized by CYP3A4 to its inactive metabolite APC. Irinotecan 63-73 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 191-197 20716241-1 2010 WHAT IS ALREADY KNOWN ABOUT THIS SUBJECT: The anticancer agent irinotecan is a prodrug that is hydrolyzed by hepatic carboxylesterase to its active and toxic metabolite SN-38 and oxidized by CYP3A4 to its inactive metabolite APC. Irinotecan 169-174 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 191-197 20716241-2 2010 Irinotecan therapy is complicated by co-administered drugs that inhibit CYP3A4 and decrease APC formation and that indirectly increase SN-38 formation. Irinotecan 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 72-78 20716241-4 2010 WHAT THIS STUDY ADDS: In microsomal fractions from patients with severe hepatic dysfunction both APC and SN-38 formation were decreased due to down-regulation of CYP3A4 and carboxylesterase enzymes. Irinotecan 105-110 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 162-168 20716241-14 2010 CONCLUSIONS: Down-regulation of CYP3A4 in liver disease decreased APC formation from irinotecan. Irinotecan 85-95 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 32-38 20716241-16 2010 However, in a subset of disease samples SN-38 production was relatively high because CYP3A4 activity was markedly impaired. Irinotecan 40-45 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 85-91 20628391-0 2010 UGT1A and TYMS genetic variants predict toxicity and response of colorectal cancer patients treated with first-line irinotecan and fluorouracil combination therapy. Irinotecan 116-126 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 0-5 20504912-4 2010 In contrast, one of its analogs, irinotecan (CPT-11), was found to be an hPXR agonist in the same tests. Irinotecan 33-43 nuclear receptor subfamily 1 group I member 2 Homo sapiens 73-77 20504912-4 2010 In contrast, one of its analogs, irinotecan (CPT-11), was found to be an hPXR agonist in the same tests. Irinotecan 45-51 nuclear receptor subfamily 1 group I member 2 Homo sapiens 73-77 21098876-12 2010 Our results indicate that rHuEpo acts via EpoR to directly inhibit DLD-1 cell growth and indirectly modulate the cytostatics effects of 5-FU and SN-38. Irinotecan 145-150 erythropoietin receptor Homo sapiens 42-46 19951251-7 2010 Exposure of melanoma cells to conditioned media from the HB1.F3.CD.rCE cells in the presence of CPT-11 increased the tumor cell-killing effect by approximately 100-fold when compared to CPT-11 alone. Irinotecan 96-102 histocompatibility minor HB-1 Homo sapiens 57-60 19951251-7 2010 Exposure of melanoma cells to conditioned media from the HB1.F3.CD.rCE cells in the presence of CPT-11 increased the tumor cell-killing effect by approximately 100-fold when compared to CPT-11 alone. Irinotecan 186-192 histocompatibility minor HB-1 Homo sapiens 57-60 20619636-3 2010 In this study, we demonstrated that enforced expression of Aven blocks UV-irradiation-, SN-38- or cisplatin-induced apoptosis upstream of mitochondria by stabilising Bcl-xL protein levels in breast cancer cells. Irinotecan 88-93 apoptosis and caspase activation inhibitor Homo sapiens 59-63 20619636-3 2010 In this study, we demonstrated that enforced expression of Aven blocks UV-irradiation-, SN-38- or cisplatin-induced apoptosis upstream of mitochondria by stabilising Bcl-xL protein levels in breast cancer cells. Irinotecan 88-93 BCL2 like 1 Homo sapiens 166-172 20066473-6 2010 We found that metronomic administration of CPT-11 significantly inhibited malignant glioma growth by inhibiting angiogenesis; this treatment procedure reduced the number of tumor vessels and the area of hypoxic lesions and reduced expression of VEGF and HIF-1alpha, the most important angiogenic factors in gliomas. Irinotecan 43-49 vascular endothelial growth factor A Homo sapiens 245-249 20066473-6 2010 We found that metronomic administration of CPT-11 significantly inhibited malignant glioma growth by inhibiting angiogenesis; this treatment procedure reduced the number of tumor vessels and the area of hypoxic lesions and reduced expression of VEGF and HIF-1alpha, the most important angiogenic factors in gliomas. Irinotecan 43-49 hypoxia inducible factor 1 subunit alpha Homo sapiens 254-264 22966396-7 2010 Among the genes, Bcl-2/adenovirus E1B 19 kDa protein interacting protein 3 (BNIP3), a Bcl-2 family pro-apoptotic protein, was identified as being involved in CPT-11 resistance following methylation of its promoter. Irinotecan 158-164 BCL2 interacting protein 3 Homo sapiens 17-74 22966396-7 2010 Among the genes, Bcl-2/adenovirus E1B 19 kDa protein interacting protein 3 (BNIP3), a Bcl-2 family pro-apoptotic protein, was identified as being involved in CPT-11 resistance following methylation of its promoter. Irinotecan 158-164 BCL2 interacting protein 3 Homo sapiens 76-81 22966396-7 2010 Among the genes, Bcl-2/adenovirus E1B 19 kDa protein interacting protein 3 (BNIP3), a Bcl-2 family pro-apoptotic protein, was identified as being involved in CPT-11 resistance following methylation of its promoter. Irinotecan 158-164 BCL2 apoptosis regulator Homo sapiens 17-22 22966396-10 2010 The results suggest that methylation of BNIP3 is a predictive factor in the prognosis and response to CPT-11 treatment in CRC patients. Irinotecan 102-108 BCL2 interacting protein 3 Homo sapiens 40-45 20620155-5 2010 The enzymatic properties of UGT1A1 proteins were characterized by kinetic analysis of 7-hydroxy-4-trifluoromethylcoumarin (7-HFC), estradiol at 3-hydroxy position (E-3OH) and 7-ethyl-10-hydroxycamptothecin (SN-38) glucuronidation. Irinotecan 175-205 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 28-34 20620155-5 2010 The enzymatic properties of UGT1A1 proteins were characterized by kinetic analysis of 7-hydroxy-4-trifluoromethylcoumarin (7-HFC), estradiol at 3-hydroxy position (E-3OH) and 7-ethyl-10-hydroxycamptothecin (SN-38) glucuronidation. Irinotecan 207-212 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 28-34 20620155-9 2010 In recombinant UGT1A1 enzymes expressed in insect cells, the kinetics of 7-HFC, E-3OH and SN-38 glucuronidation fitted the substrate inhibition (7-HFC glucuronidation) or Hill equation (E-3OH and SN-38 glucuronidation), and each glucuronidation showed the same kinetic profile between humans and cynomolgus monkeys. Irinotecan 90-95 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 15-21 20628391-9 2010 CONCLUSION: TYMS and UGT1A polymorphisms influence on tumour response and toxicities derived from irinotecan/5FU treatment in CRC patients. Irinotecan 98-108 thymidylate synthetase Homo sapiens 12-16 20628391-9 2010 CONCLUSION: TYMS and UGT1A polymorphisms influence on tumour response and toxicities derived from irinotecan/5FU treatment in CRC patients. Irinotecan 98-108 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 21-26 20628391-0 2010 UGT1A and TYMS genetic variants predict toxicity and response of colorectal cancer patients treated with first-line irinotecan and fluorouracil combination therapy. Irinotecan 116-126 thymidylate synthetase Homo sapiens 10-14 20666740-0 2010 Prospective evaluation of angiogenic, hypoxic and EGFR-related biomarkers in recurrent glioblastoma multiforme treated with cetuximab, bevacizumab and irinotecan. Irinotecan 151-161 epidermal growth factor receptor Homo sapiens 50-54 20649590-1 2010 BACKGROUND AND PURPOSE: Carboxylesterases (CEs) metabolize a wide range of xenobiotic substrates including heroin, cocaine, meperidine and the anticancer agent CPT-11. Irinotecan 160-166 carboxylesterase 1 Homo sapiens 24-41 20649590-7 2010 However, cocaine, heroin and CPT-11 were all substrates for the intestinal CE, hiCE (CES2), as determined using both the recombinant protein and the tissue fractions. Irinotecan 29-35 carboxylesterase 2 Homo sapiens 85-89 20653675-0 2010 Association of carboxylesterase 1A genotypes with irinotecan pharmacokinetics in Japanese cancer patients. Irinotecan 50-60 carboxylesterase 1 Homo sapiens 15-33 20406168-5 2010 In recent years, genetic polymorphisms in UDP-glucuronosyltransferase (UGT) 1A1, an enzyme involved in SN-38 glucuronidation, has been linked to interindividual pharmacokinetic variability and irinotecan toxicity. Irinotecan 103-108 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 42-79 20653675-1 2010 WHAT IS ALREADY KNOWN ABOUT THIS SUBJECT * Association of UDP-glucuronosyltransferase 1A1 (UGT1A1) genetic polymorphisms *6 and *28 with reduced clearance of SN-38 and severe neutropenia in irinotecan therapy was demonstrated in Japanese cancer patients. Irinotecan 158-163 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 58-89 20653675-1 2010 WHAT IS ALREADY KNOWN ABOUT THIS SUBJECT * Association of UDP-glucuronosyltransferase 1A1 (UGT1A1) genetic polymorphisms *6 and *28 with reduced clearance of SN-38 and severe neutropenia in irinotecan therapy was demonstrated in Japanese cancer patients. Irinotecan 158-163 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 91-97 20653675-1 2010 WHAT IS ALREADY KNOWN ABOUT THIS SUBJECT * Association of UDP-glucuronosyltransferase 1A1 (UGT1A1) genetic polymorphisms *6 and *28 with reduced clearance of SN-38 and severe neutropenia in irinotecan therapy was demonstrated in Japanese cancer patients. Irinotecan 190-200 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 58-89 20653675-1 2010 WHAT IS ALREADY KNOWN ABOUT THIS SUBJECT * Association of UDP-glucuronosyltransferase 1A1 (UGT1A1) genetic polymorphisms *6 and *28 with reduced clearance of SN-38 and severe neutropenia in irinotecan therapy was demonstrated in Japanese cancer patients. Irinotecan 190-200 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 91-97 20653675-4 2010 WHAT THIS STUDY ADDS * Association of functional CES1 gene number with AUC ratio [(SN-38 + SN-38G)/irinotecan], an in vivo index of CES activity, was observed in patients with irinotecan monotherapy. Irinotecan 83-88 carboxylesterase 1 Homo sapiens 49-53 20653675-4 2010 WHAT THIS STUDY ADDS * Association of functional CES1 gene number with AUC ratio [(SN-38 + SN-38G)/irinotecan], an in vivo index of CES activity, was observed in patients with irinotecan monotherapy. Irinotecan 99-109 carboxylesterase 1 Homo sapiens 49-53 20653675-4 2010 WHAT THIS STUDY ADDS * Association of functional CES1 gene number with AUC ratio [(SN-38 + SN-38G)/irinotecan], an in vivo index of CES activity, was observed in patients with irinotecan monotherapy. Irinotecan 176-186 carboxylesterase 1 Homo sapiens 49-53 20653675-6 2010 AIMS Human carboxylesterase 1 (CES1) hydrolyzes irinotecan to produce an active metabolite SN-38 in the liver. Irinotecan 48-58 carboxylesterase 1 Homo sapiens 11-29 20653675-6 2010 AIMS Human carboxylesterase 1 (CES1) hydrolyzes irinotecan to produce an active metabolite SN-38 in the liver. Irinotecan 48-58 carboxylesterase 1 Homo sapiens 31-35 20653675-6 2010 AIMS Human carboxylesterase 1 (CES1) hydrolyzes irinotecan to produce an active metabolite SN-38 in the liver. Irinotecan 91-96 carboxylesterase 1 Homo sapiens 11-29 20653675-6 2010 AIMS Human carboxylesterase 1 (CES1) hydrolyzes irinotecan to produce an active metabolite SN-38 in the liver. Irinotecan 91-96 carboxylesterase 1 Homo sapiens 31-35 20653675-11 2010 Associations of CES1 genotypes, number of functional CES1 genes (1A1, 1A2 and var1A1) and major SNPs, with the AUC ratio of (SN-38 + SN-38G)/irinotecan, a parameter of in vivo CES activity, were analyzed for 58 patients treated with irinotecan monotherapy. Irinotecan 125-130 carboxylesterase 1 Homo sapiens 16-20 20653675-11 2010 Associations of CES1 genotypes, number of functional CES1 genes (1A1, 1A2 and var1A1) and major SNPs, with the AUC ratio of (SN-38 + SN-38G)/irinotecan, a parameter of in vivo CES activity, were analyzed for 58 patients treated with irinotecan monotherapy. Irinotecan 141-151 carboxylesterase 1 Homo sapiens 16-20 20562211-0 2010 Dose-dependent association between UGT1A1*28 genotype and irinotecan-induced neutropenia: low doses also increase risk. Irinotecan 58-68 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 35-41 20562211-1 2010 PURPOSE: A previous meta-analysis showed that the association between the UGT1A1*28 genotype and irinotecan-induced neutropenia was influenced by irinotecan dose and that the risk of neutropenia was similar at low doses for patients with all genotypes. Irinotecan 97-107 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 74-80 20562211-1 2010 PURPOSE: A previous meta-analysis showed that the association between the UGT1A1*28 genotype and irinotecan-induced neutropenia was influenced by irinotecan dose and that the risk of neutropenia was similar at low doses for patients with all genotypes. Irinotecan 146-156 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 74-80 20562211-5 2010 The association between UGT1A1*28 and the relative extent of glucuronidation (REG) of SN-38 was also examined. Irinotecan 86-91 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 24-30 20562211-5 2010 The association between UGT1A1*28 and the relative extent of glucuronidation (REG) of SN-38 was also examined. Irinotecan 86-91 regenerating family member 1 alpha Homo sapiens 78-81 20562211-11 2010 We found the weighted mean difference of REG (UGT1A1*1/*1 or UGT1A1*1/*28 versus UGT1A1*28/*28) increased with increasing dose of irinotecan. Irinotecan 130-140 regenerating family member 1 alpha Homo sapiens 41-44 20562211-11 2010 We found the weighted mean difference of REG (UGT1A1*1/*1 or UGT1A1*1/*28 versus UGT1A1*28/*28) increased with increasing dose of irinotecan. Irinotecan 130-140 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 46-52 20562211-11 2010 We found the weighted mean difference of REG (UGT1A1*1/*1 or UGT1A1*1/*28 versus UGT1A1*28/*28) increased with increasing dose of irinotecan. Irinotecan 130-140 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 61-67 20562211-11 2010 We found the weighted mean difference of REG (UGT1A1*1/*1 or UGT1A1*1/*28 versus UGT1A1*28/*28) increased with increasing dose of irinotecan. Irinotecan 130-140 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 61-67 20562211-12 2010 CONCLUSIONS: In conclusion, the UGT1A1*28/*28 genotype was associated with an increased risk of neutropenia not only at medium or high doses of irinotecan but also at low doses. Irinotecan 144-154 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 32-38 20562211-13 2010 The dose-dependent manner of SN-38 glucuronidation explained why the association between UGT1A1*28 and neutropenia was dose dependent. Irinotecan 29-34 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 89-95 20406168-5 2010 In recent years, genetic polymorphisms in UDP-glucuronosyltransferase (UGT) 1A1, an enzyme involved in SN-38 glucuronidation, has been linked to interindividual pharmacokinetic variability and irinotecan toxicity. Irinotecan 193-203 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 42-79 20580994-0 2010 The UGT1A1*28 polymorphism correlates with erlotinib"s effect on SN-38 glucuronidation. Irinotecan 65-70 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 20663031-3 2010 In addition, we have investigated the potential involvement of breast cancer resistance protein (BCRP) in biliary excretion, pharmacokinetics, and intestinal toxicity of CPT-11. Irinotecan 170-176 ATP binding cassette subfamily G member 2 Canis lupus familiaris 97-101 20663031-5 2010 The effect of PEO-PPO-PEO micelles on BCRP-mediated cellular accumulation and transport efflux of CPT-11 was evaluated in MDCKII/BCRP cells. Irinotecan 98-104 ATP binding cassette subfamily G member 2 Canis lupus familiaris 38-42 20663031-5 2010 The effect of PEO-PPO-PEO micelles on BCRP-mediated cellular accumulation and transport efflux of CPT-11 was evaluated in MDCKII/BCRP cells. Irinotecan 98-104 ATP binding cassette subfamily G member 2 Canis lupus familiaris 129-133 20663031-7 2010 KEY FINDINGS: The obtained micelles could effectively inhibit BCRP-mediated CPT-11 efflux in MDCKII/BCRP cells, and significantly decrease the drug biliary excretion in rats. Irinotecan 76-82 ATP binding cassette subfamily G member 2 Canis lupus familiaris 62-66 20663031-7 2010 KEY FINDINGS: The obtained micelles could effectively inhibit BCRP-mediated CPT-11 efflux in MDCKII/BCRP cells, and significantly decrease the drug biliary excretion in rats. Irinotecan 76-82 ATP binding cassette subfamily G member 2 Canis lupus familiaris 100-104 20663031-11 2010 The study indicated a potential involvement of BCRP in CPT-11 pharmacokinetics and CPT-11-induced intestinal toxicity. Irinotecan 55-61 ATP binding cassette subfamily G member 2 Canis lupus familiaris 47-51 20663031-11 2010 The study indicated a potential involvement of BCRP in CPT-11 pharmacokinetics and CPT-11-induced intestinal toxicity. Irinotecan 83-89 ATP binding cassette subfamily G member 2 Canis lupus familiaris 47-51 20661248-1 2010 BACKGROUND: Accurate description of current practice within advanced colorectal cancer (CRC) specialties were needed to inform an economic evaluation of the UGT1A1 pharmacogenetic test for irinotecan in the United Kingdom. Irinotecan 189-199 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 157-163 20637356-4 2010 The phenotyping of DPD and UGT1A1 activities, and to a lesser extent, its genotyping, appears as the most useful tool in terms of prediction of toxicities induced by two major drugs: 5-FU and irinotecan. Irinotecan 192-202 dihydropyrimidine dehydrogenase Homo sapiens 19-22 20637356-4 2010 The phenotyping of DPD and UGT1A1 activities, and to a lesser extent, its genotyping, appears as the most useful tool in terms of prediction of toxicities induced by two major drugs: 5-FU and irinotecan. Irinotecan 192-202 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 27-33 20630823-2 2010 This trial was designed to evaluate irinotecan/cisplatin plus maintenance imatinib in patients with c-Kit-positive disease (the transmembrane receptor c-Kit is the product of the c-KIT protooncogene). Irinotecan 36-46 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 100-105 20630823-2 2010 This trial was designed to evaluate irinotecan/cisplatin plus maintenance imatinib in patients with c-Kit-positive disease (the transmembrane receptor c-Kit is the product of the c-KIT protooncogene). Irinotecan 36-46 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 151-156 20630823-2 2010 This trial was designed to evaluate irinotecan/cisplatin plus maintenance imatinib in patients with c-Kit-positive disease (the transmembrane receptor c-Kit is the product of the c-KIT protooncogene). Irinotecan 36-46 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 179-184 20063115-3 2010 The MTD for patients receiving enzyme-inducing anticonvulsants (EIAC) was as follows: irinotecan 400 mg/m(2)/week on Days 1, 8, 22 and 29 during RT, followed by BCNU 100 mg/m(2) Day 1, and irinotecan, 400 mg/m(2) on Days 1, 8, 22 and 29, every 6 weeks. Irinotecan 86-96 metallothionein 1E Homo sapiens 4-7 20063115-10 2010 SN-38 C(max) and AUC in EIAC patients receiving 400 mg/m(2) irinotecan were 20.9 ng/ml and 212 ng/ml h, and in non-EIAC patients receiving 125 mg/m(2), 15.5 ng/ml and 207 ng/ml h. SN-38 AUC varied by UGT1A1*28 status in non-EIAC patients. Irinotecan 0-5 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 200-206 20063115-12 2010 Non-EIAC patients with UGT1A1*28 variant alleles appear particularly sensitive to toxicity from irinotecan. Irinotecan 96-106 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 23-29 20580994-2 2010 We aimed to investigate the effect of erlotinib on SN-38 glucuronidation and the association between UGT1A polymorphisms and SN-38 glucuronidation activity in the presence of erlotinib. Irinotecan 125-130 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 101-106 20580994-6 2010 Our data showed that erlotinib exhibited potent non-competitive inhibition against SN-38 glucuronidation in pooled HLMs and UGT1A1. Irinotecan 83-88 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 124-130 20580994-9 2010 Significant correlations were observed between SN-38 glucuronidation activity in the presence of erlotinib and UGT1A1*28 in 52 Caucasian liver microsomes. Irinotecan 47-52 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 111-117 20580994-10 2010 Our results suggest that erlotinib is a potent inhibitor of SN-38 glucuronidation via UGT1A1 inhibition. Irinotecan 60-65 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 86-92 20580994-12 2010 UGT1A1*28 polymorphism correlates with erlotinib"s effect on SN-38 glucuronidation. Irinotecan 61-66 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 19739070-2 2010 We investigated the influence of the essential component of MMR, the hMLH1 protein, on the cellular response to DNA-double strand breaks induced by treatment with SN-38, the active metabolite of topoisomerase I inhibitor irinotecan, in a strictly isogenic cell system (p53(wt), hMLH1(+)/p53(wt), hMLH1(-)). Irinotecan 163-168 mutL homolog 1 Homo sapiens 69-74 20335017-0 2010 Dose-dependent association between UGT1A1*28 polymorphism and irinotecan-induced diarrhoea: a meta-analysis. Irinotecan 62-72 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 35-41 20335017-2 2010 The associations between the UGT1A1*28 polymorphism and irinotecan-induced diarrhoea remains controversial because of conflicting data in the literature. Irinotecan 56-66 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 29-35 20304778-0 2010 Phase II study of combination chemotherapy with biweekly cetuximab and irinotecan for pre-treated metastatic colorectal cancer harboring wild-type KRAS. Irinotecan 71-81 KRAS proto-oncogene, GTPase Homo sapiens 147-151 20602618-3 2010 Although multiple genes may play a role in irinotecan activity, the majority of evidence to date suggests that variation in expression of UGT1A1 caused by a common promoter polymorphism (UGT1A1*28) is strongly associated with toxicity; however, this link is dose dependent. Irinotecan 43-53 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 138-144 20602618-3 2010 Although multiple genes may play a role in irinotecan activity, the majority of evidence to date suggests that variation in expression of UGT1A1 caused by a common promoter polymorphism (UGT1A1*28) is strongly associated with toxicity; however, this link is dose dependent. Irinotecan 43-53 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 187-193 20602618-4 2010 Variations in other pharmacokinetic genes, particularly the transporter ABCC2, also contribute to irinotecan toxicity. Irinotecan 98-108 ATP binding cassette subfamily C member 2 Homo sapiens 72-77 20335017-9 2010 The risk of severe diarrhoea at medium and high irinotecan doses was higher among patients with a UGT1A1*28/*28 genotype than among those with a UGT1A1*1/*1 genotype (OR=3.69, 95% confidence interval [CI]=2.00-6.83; P<0.001). Irinotecan 48-58 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 98-104 20335017-12 2010 In conclusion, patients carrying UGT1A1*28 allele(s) are at an increased risk of irinotecan-induced severe diarrhoea. Irinotecan 81-91 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-39 19739070-3 2010 By using hMLH1 expressing clones or cells transduced with the hMLH1-expressing adenovirus as well as siRNA technology, we show that in response to SN-38-induced DNA damage the MMR proficient (MMR(+)) cells make: (i) a stronger G2/M arrest, (ii) a subsequent longer tetraploid G1 arrest, (iii) a stronger activation of Chk1 and Chk2 kinases than the MMR deficient (MMR(-)) counterparts. Irinotecan 147-152 mutL homolog 1 Homo sapiens 9-14 19739070-3 2010 By using hMLH1 expressing clones or cells transduced with the hMLH1-expressing adenovirus as well as siRNA technology, we show that in response to SN-38-induced DNA damage the MMR proficient (MMR(+)) cells make: (i) a stronger G2/M arrest, (ii) a subsequent longer tetraploid G1 arrest, (iii) a stronger activation of Chk1 and Chk2 kinases than the MMR deficient (MMR(-)) counterparts. Irinotecan 147-152 mutL homolog 1 Homo sapiens 62-67 19739070-3 2010 By using hMLH1 expressing clones or cells transduced with the hMLH1-expressing adenovirus as well as siRNA technology, we show that in response to SN-38-induced DNA damage the MMR proficient (MMR(+)) cells make: (i) a stronger G2/M arrest, (ii) a subsequent longer tetraploid G1 arrest, (iii) a stronger activation of Chk1 and Chk2 kinases than the MMR deficient (MMR(-)) counterparts. Irinotecan 147-152 checkpoint kinase 1 Homo sapiens 318-322 19739070-3 2010 By using hMLH1 expressing clones or cells transduced with the hMLH1-expressing adenovirus as well as siRNA technology, we show that in response to SN-38-induced DNA damage the MMR proficient (MMR(+)) cells make: (i) a stronger G2/M arrest, (ii) a subsequent longer tetraploid G1 arrest, (iii) a stronger activation of Chk1 and Chk2 kinases than the MMR deficient (MMR(-)) counterparts. Irinotecan 147-152 checkpoint kinase 2 Homo sapiens 327-331 19739070-2 2010 We investigated the influence of the essential component of MMR, the hMLH1 protein, on the cellular response to DNA-double strand breaks induced by treatment with SN-38, the active metabolite of topoisomerase I inhibitor irinotecan, in a strictly isogenic cell system (p53(wt), hMLH1(+)/p53(wt), hMLH1(-)). Irinotecan 221-231 mutL homolog 1 Homo sapiens 69-74 20124398-3 2010 The well characterized substrate for UGT1A1, 7-ethyl-10-hydroxy-camptothecin (SN-38), showed the greatest difference in parent drug exposure ( approximately 3-fold increase) and clearance ( approximately 3-fold decrease) in Tg(UGT1(A1*28)) Ugt1(-/-) mice after intravenous administration compared with wild-type and phenobarbital-treated animals. Irinotecan 78-83 UDP glucuronosyltransferase 1 family, polypeptide A1 Mus musculus 37-43 19908066-11 2010 CONCLUSIONS: XRCC1 genotype independently predicted overall survival in metastatic colorectal carcinoma patients treated with irinotecan-based chemotherapy. Irinotecan 126-136 X-ray repair cross complementing 1 Homo sapiens 13-18 20211505-1 2010 BACKGROUND AND PURPOSE: The aim of this study is to evaluate the efficacy and safety of preoperative radiation therapy combined with S-1 and irinotecan (SI) in LARC. Irinotecan 141-151 C-C motif chemokine ligand 20 Homo sapiens 160-164 20121626-0 2010 Carcinoembryonic antigen surge in metastatic colorectal cancer patients responding to irinotecan combination chemotherapy. Irinotecan 86-96 CEA cell adhesion molecule 3 Homo sapiens 0-24 20121626-2 2010 The aim of this study was to investigate the phenomenon of CEA surge in irinotecan-based chemotherapy. Irinotecan 72-82 CEA cell adhesion molecule 3 Homo sapiens 59-62 20121626-7 2010 CONCLUSION: A CEA surge can be induced by irinotecan-based chemotherapy. Irinotecan 42-52 CEA cell adhesion molecule 3 Homo sapiens 14-17 20121626-8 2010 An early increase in CEA after irinotecan-based chemotherapy does not usually indicate progression of disease and failure of therapy, and should not lead to a change of chemotherapy. Irinotecan 31-41 CEA cell adhesion molecule 3 Homo sapiens 21-24 20164124-0 2010 Secreted protein acidic and rich in cysteine-induced cellular senescence in colorectal cancers in response to irinotecan is mediated by P53. Irinotecan 110-120 secreted protein acidic and cysteine rich Homo sapiens 0-44 20164124-0 2010 Secreted protein acidic and rich in cysteine-induced cellular senescence in colorectal cancers in response to irinotecan is mediated by P53. Irinotecan 110-120 tumor protein p53 Homo sapiens 136-139 20164124-6 2010 However, CPT-11-resistant cells exposed to endogenous or exogenous SPARC can also be triggered into cellular senescence. Irinotecan 9-15 secreted protein acidic and cysteine rich Homo sapiens 67-72 20651349-12 2010 Silencing of beta-catenin decreased transcription of these ABC transporter genes; beta-catenin-silenced cells became relatively sensitive to paclitaxel and irinotecan. Irinotecan 156-166 catenin beta 1 Homo sapiens 13-25 20651349-12 2010 Silencing of beta-catenin decreased transcription of these ABC transporter genes; beta-catenin-silenced cells became relatively sensitive to paclitaxel and irinotecan. Irinotecan 156-166 catenin beta 1 Homo sapiens 82-94 19771428-2 2010 METHODS: Associations between transporter haplotypes/variations along with UGT1A1*28 or *6 and SN-38 area under the time-concentration curve (AUC) or neutropenia were examined in irinotecan monotherapy (55 patients) and irinotecan-cisplatin-combination therapy (62 patients). Irinotecan 95-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 75-81 19771428-3 2010 RESULTS: Higher SN-38 AUC values were observed in ABCB1 2677G>T (A893S) (*2 group) for both regimens. Irinotecan 16-21 ATP binding cassette subfamily B member 1 Homo sapiens 50-55 20124398-8 2010 These differences are consistent with SN-38 glucuronidation activities using HLMs isolated from individuals genotyped as UGT1A1*1 or UGT1A1*28. Irinotecan 38-43 UDP glucuronosyltransferase 1 family, polypeptide A1 Mus musculus 121-127 20124398-3 2010 The well characterized substrate for UGT1A1, 7-ethyl-10-hydroxy-camptothecin (SN-38), showed the greatest difference in parent drug exposure ( approximately 3-fold increase) and clearance ( approximately 3-fold decrease) in Tg(UGT1(A1*28)) Ugt1(-/-) mice after intravenous administration compared with wild-type and phenobarbital-treated animals. Irinotecan 78-83 UDP glucuronosyltransferase 1 family, polypeptide A2 Mus musculus 37-41 20124398-3 2010 The well characterized substrate for UGT1A1, 7-ethyl-10-hydroxy-camptothecin (SN-38), showed the greatest difference in parent drug exposure ( approximately 3-fold increase) and clearance ( approximately 3-fold decrease) in Tg(UGT1(A1*28)) Ugt1(-/-) mice after intravenous administration compared with wild-type and phenobarbital-treated animals. Irinotecan 78-83 UDP glucuronosyltransferase 1 family, polypeptide A2 Mus musculus 240-244 20124398-7 2010 For SN-38 glucuronidation, V(max) decreased 5-fold in Tg(UGT1(A1*28)) Ugt1(-/-) mouse liver microsomes compared with microsomes prepared from wild-type mice, and decreased 10-fold compared with phenobarbital-treated Tg(UGT1(A1*28)) Ugt1(-/-) mice. Irinotecan 4-9 UDP glucuronosyltransferase 1 family, polypeptide A2 Mus musculus 57-61 20124398-7 2010 For SN-38 glucuronidation, V(max) decreased 5-fold in Tg(UGT1(A1*28)) Ugt1(-/-) mouse liver microsomes compared with microsomes prepared from wild-type mice, and decreased 10-fold compared with phenobarbital-treated Tg(UGT1(A1*28)) Ugt1(-/-) mice. Irinotecan 4-9 UDP glucuronosyltransferase 1 family, polypeptide A2 Mus musculus 70-74 20124398-7 2010 For SN-38 glucuronidation, V(max) decreased 5-fold in Tg(UGT1(A1*28)) Ugt1(-/-) mouse liver microsomes compared with microsomes prepared from wild-type mice, and decreased 10-fold compared with phenobarbital-treated Tg(UGT1(A1*28)) Ugt1(-/-) mice. Irinotecan 4-9 UDP glucuronosyltransferase 1 family, polypeptide A2 Mus musculus 219-223 20124398-7 2010 For SN-38 glucuronidation, V(max) decreased 5-fold in Tg(UGT1(A1*28)) Ugt1(-/-) mouse liver microsomes compared with microsomes prepared from wild-type mice, and decreased 10-fold compared with phenobarbital-treated Tg(UGT1(A1*28)) Ugt1(-/-) mice. Irinotecan 4-9 UDP glucuronosyltransferase 1 family, polypeptide A2 Mus musculus 232-236 20085586-8 2010 Patients that were homozygous for the reference alleles SSTR4 rs2567608 (AA) and EPHA7 rs2278107 (TT) showed lower disease control rates in response to irinotecan and oxaliplatin regimens, respectively, than those with substitution alleles (P = 0.022 and 0.014, respectively). Irinotecan 152-162 somatostatin receptor 4 Homo sapiens 56-61 20395318-2 2010 We investigated the kinetics of (18)F-FLT during treatment of malignant glioma with bevacizumab and irinotecan. Irinotecan 100-110 fms related receptor tyrosine kinase 1 Homo sapiens 38-41 20371676-0 2010 Aprataxin tumor levels predict response of colorectal cancer patients to irinotecan-based treatment. Irinotecan 73-83 aprataxin Homo sapiens 0-9 20371676-4 2010 RESULTS: A good correlation was observed between irinotecan sensitivity and the expression of aprataxin (APTX), a histidine triad domain superfamily protein involved in DNA repair. Irinotecan 49-59 aprataxin Homo sapiens 94-103 20371676-4 2010 RESULTS: A good correlation was observed between irinotecan sensitivity and the expression of aprataxin (APTX), a histidine triad domain superfamily protein involved in DNA repair. Irinotecan 49-59 aprataxin Homo sapiens 105-109 20371676-5 2010 Moreover, using an isogenic in vitro system deficient in APTX, we show that aprataxin directly regulates the cellular sensitivity to camptothecin, suggesting that it could be used to predict patient response to irinotecan. Irinotecan 211-221 aprataxin Homo sapiens 76-85 20371676-10 2010 CONCLUSIONS: These results show that aprataxin tumor levels can be used to identify patients with low probability of response to irinotecan-based therapy who are ideal candidates to receive treatment with alternative agents in an attempt to improve patient survival. Irinotecan 129-139 aprataxin Homo sapiens 37-46 20513938-3 2010 Patients with genetic variations in UGT1A1 and DPYD genes are hypersensitive to Irinotecan and 5-Fluorouracil (5FU) respectively. Irinotecan 80-90 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 36-42 20513938-3 2010 Patients with genetic variations in UGT1A1 and DPYD genes are hypersensitive to Irinotecan and 5-Fluorouracil (5FU) respectively. Irinotecan 80-90 dihydropyrimidine dehydrogenase Homo sapiens 47-51 20530432-2 2010 MATERIALS AND METHODS: We evaluated the influence of hepatocyte growth factor, vascular endothelial growth factor and epidermal growth factor on the effect of 5-fluorouracil, oxaliplatin and SN-38 (the active metabolite of irinotecan) on WiDr cells. Irinotecan 191-196 vascular endothelial growth factor A Homo sapiens 79-113 20530432-2 2010 MATERIALS AND METHODS: We evaluated the influence of hepatocyte growth factor, vascular endothelial growth factor and epidermal growth factor on the effect of 5-fluorouracil, oxaliplatin and SN-38 (the active metabolite of irinotecan) on WiDr cells. Irinotecan 191-196 epidermal growth factor Homo sapiens 118-141 20085586-8 2010 Patients that were homozygous for the reference alleles SSTR4 rs2567608 (AA) and EPHA7 rs2278107 (TT) showed lower disease control rates in response to irinotecan and oxaliplatin regimens, respectively, than those with substitution alleles (P = 0.022 and 0.014, respectively). Irinotecan 152-162 EPH receptor A7 Homo sapiens 81-86 20704575-5 2010 Combined treatments with CPT-11 or SN-38 significantly increased caspase 3/7 activity compared with RET siRNA, CPT-11 or SN-38 treatment alone. Irinotecan 25-31 caspase 3 Homo sapiens 65-74 20047821-7 2010 In comparison with bare CS-g-PCL nanomicelles, the corresponding SN-38-loaded nanomicelles showed increased particle sizes and a little reduced zeta potentials. Irinotecan 65-70 polycystic kidney disease 2-like 1 Mus musculus 29-32 20704575-5 2010 Combined treatments with CPT-11 or SN-38 significantly increased caspase 3/7 activity compared with RET siRNA, CPT-11 or SN-38 treatment alone. Irinotecan 25-31 ret proto-oncogene Homo sapiens 100-103 20704575-5 2010 Combined treatments with CPT-11 or SN-38 significantly increased caspase 3/7 activity compared with RET siRNA, CPT-11 or SN-38 treatment alone. Irinotecan 35-40 caspase 3 Homo sapiens 65-74 20704575-5 2010 Combined treatments with CPT-11 or SN-38 significantly increased caspase 3/7 activity compared with RET siRNA, CPT-11 or SN-38 treatment alone. Irinotecan 35-40 ret proto-oncogene Homo sapiens 100-103 20704575-7 2010 These findings that RET siRNA enhanced sensitivity for CPT-11 will provide a novel strategy for the treatment of MTC with RET mutation. Irinotecan 55-61 ret proto-oncogene Homo sapiens 20-23 20704575-7 2010 These findings that RET siRNA enhanced sensitivity for CPT-11 will provide a novel strategy for the treatment of MTC with RET mutation. Irinotecan 55-61 ret proto-oncogene Homo sapiens 122-125 20047821-6 2010 Water-insoluble antitumor drug, SN-38, was easily encapsulated into CS-g-PCL nanomicelles by lyophilization method. Irinotecan 32-37 polycystic kidney disease 2-like 1 Mus musculus 73-76 20047821-9 2010 Also, the CS-g-PCL nanomicelles effectively protected the active lactone ring of SN-38 from hydrolysis under physiological condition, due to the encapsulation of SN-38 into the hydrophobic cores in the nanomicelles. Irinotecan 81-86 polycystic kidney disease 2-like 1 Mus musculus 15-18 20047821-9 2010 Also, the CS-g-PCL nanomicelles effectively protected the active lactone ring of SN-38 from hydrolysis under physiological condition, due to the encapsulation of SN-38 into the hydrophobic cores in the nanomicelles. Irinotecan 162-167 polycystic kidney disease 2-like 1 Mus musculus 15-18 20511137-3 2010 Numerous variants have been found in UGT1A1 , the main conjugating enzyme of bilirubin and drugs such as the anticancer drug irinotecan. Irinotecan 125-135 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 37-43 20511137-4 2010 Recently, the US Food and Drug Administration (FDA) recommended testing for the presence of UGT1A1*28 , an allele correlated with decreased transcriptional activity, to predict patients at risk of irinotecan toxicity. Irinotecan 197-207 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 92-98 20179984-0 2010 Indispensability of UGT1A1*6 genotyping in Japanese cancer patients treated with irinotecan. Irinotecan 81-91 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 20-26 20726333-7 2010 In cocultures, IL-1beta induced HGF release, while CPT-11 alone or combined with IL-1beta decreased HGF secretion. Irinotecan 51-57 hepatocyte growth factor Homo sapiens 100-103 20726333-8 2010 An immunoblotting analysis followed by densitometry revealed that the combination of IL-1beta plus CPT-11 added to the cocultures led to a decrease in HSPs and MRP levels. Irinotecan 99-105 ATP binding cassette subfamily C member 1 Homo sapiens 160-163 19859084-0 2010 Association of SUMO1 and UBC9 genotypes with tumor response in non-small-cell lung cancer treated with irinotecan-based chemotherapy. Irinotecan 103-113 small ubiquitin like modifier 1 Homo sapiens 15-20 20216081-3 2010 The purpose of this study is to determine the toxicity and response rate to the addition of the selective oral COX-2 inhibitor, celecoxib, to gemcitabine and irinotecan in patients with inoperable pancreatic cancer. Irinotecan 158-168 prostaglandin-endoperoxide synthase 2 Homo sapiens 111-116 19859084-0 2010 Association of SUMO1 and UBC9 genotypes with tumor response in non-small-cell lung cancer treated with irinotecan-based chemotherapy. Irinotecan 103-113 ubiquitin conjugating enzyme E2 I Homo sapiens 25-29 19859084-1 2010 Irinotecan induces small ubiquitin-like modifier (SUMO)-1 conjugation to topoisomerase-I, leading to enhanced sensitivity to irinotecan. Irinotecan 0-10 small ubiquitin like modifier 1 Homo sapiens 19-57 19859084-1 2010 Irinotecan induces small ubiquitin-like modifier (SUMO)-1 conjugation to topoisomerase-I, leading to enhanced sensitivity to irinotecan. Irinotecan 125-135 small ubiquitin like modifier 1 Homo sapiens 19-57 19859084-7 2010 This finding suggests that the UBC9 10920CG genotype enhances sensitivity to irinotecan chemotherapy in advanced NSCLC through upregulation of SUMO1 in tumor cells. Irinotecan 77-87 ubiquitin conjugating enzyme E2 I Homo sapiens 31-35 19859084-7 2010 This finding suggests that the UBC9 10920CG genotype enhances sensitivity to irinotecan chemotherapy in advanced NSCLC through upregulation of SUMO1 in tumor cells. Irinotecan 77-87 small ubiquitin like modifier 1 Homo sapiens 143-148 20213647-3 2010 We adapted an "in vivo complexes of enzyme to DNA" (ICE) bioassay to assess irinotecan activity in the stomach, duodenum, colon and liver of male Wistar rats after a single treatment with irinotecan (100 mg/kg body weight, intraperitoneally). Irinotecan 188-198 caspase 1 Rattus norvegicus 52-55 20238327-19 2010 Two and three-drug regimens including 5-FU, cisplatin, with or without an anthracycline, as well as irinotecan or docetaxel-containing regimens are reasonable treatment options for HER-2 negative patients. Irinotecan 100-110 erb-b2 receptor tyrosine kinase 2 Homo sapiens 181-186 20196838-0 2010 Pregnane X Receptor (PXR) expression in colorectal cancer cells restricts irinotecan chemosensitivity through enhanced SN-38 glucuronidation. Irinotecan 74-84 nuclear receptor subfamily 1 group I member 2 Homo sapiens 0-19 20196838-0 2010 Pregnane X Receptor (PXR) expression in colorectal cancer cells restricts irinotecan chemosensitivity through enhanced SN-38 glucuronidation. Irinotecan 74-84 nuclear receptor subfamily 1 group I member 2 Homo sapiens 21-24 20196838-0 2010 Pregnane X Receptor (PXR) expression in colorectal cancer cells restricts irinotecan chemosensitivity through enhanced SN-38 glucuronidation. Irinotecan 119-124 nuclear receptor subfamily 1 group I member 2 Homo sapiens 0-19 20196838-0 2010 Pregnane X Receptor (PXR) expression in colorectal cancer cells restricts irinotecan chemosensitivity through enhanced SN-38 glucuronidation. Irinotecan 119-124 nuclear receptor subfamily 1 group I member 2 Homo sapiens 21-24 20196838-4 2010 Considering the metabolic pathway of irinotecan and the tissue distribution of Pregnane x Receptor (PXR), we hypothesized that PXR could play a key role in colon cancer cell response to irinotecan. Irinotecan 37-47 nuclear receptor subfamily 1 group I member 2 Homo sapiens 127-130 20196838-4 2010 Considering the metabolic pathway of irinotecan and the tissue distribution of Pregnane x Receptor (PXR), we hypothesized that PXR could play a key role in colon cancer cell response to irinotecan. Irinotecan 186-196 nuclear receptor subfamily 1 group I member 2 Homo sapiens 79-98 20196838-4 2010 Considering the metabolic pathway of irinotecan and the tissue distribution of Pregnane x Receptor (PXR), we hypothesized that PXR could play a key role in colon cancer cell response to irinotecan. Irinotecan 186-196 nuclear receptor subfamily 1 group I member 2 Homo sapiens 100-103 20196838-4 2010 Considering the metabolic pathway of irinotecan and the tissue distribution of Pregnane x Receptor (PXR), we hypothesized that PXR could play a key role in colon cancer cell response to irinotecan. Irinotecan 186-196 nuclear receptor subfamily 1 group I member 2 Homo sapiens 127-130 20196838-7 2010 Cellular response to irinotecan and its active metabolic SN38 was assessed by cell viability assays, HPLC metabolic profiles and mRNA quantification of PXR target genes. Irinotecan 21-31 nuclear receptor subfamily 1 group I member 2 Homo sapiens 152-155 20196838-13 2010 CONCLUSION: Our results demonstrate that tumoral metabolism of SN38 is affected by PXR and point to potential therapeutic significance of PXR quantification in the prediction of irinotecan response. Irinotecan 178-188 nuclear receptor subfamily 1 group I member 2 Homo sapiens 138-141 20009028-8 2010 When gavaged with fat (trioleate), RS4 stimulated a lower postprandial glucose-dependent insulinotropic polypeptide (GIP; incretin) response than RS2. Irinotecan 35-38 gastric inhibitory polypeptide Mus musculus 71-115 20009028-8 2010 When gavaged with fat (trioleate), RS4 stimulated a lower postprandial glucose-dependent insulinotropic polypeptide (GIP; incretin) response than RS2. Irinotecan 35-38 gastric inhibitory polypeptide Mus musculus 117-120 20009028-10 2010 In conclusion, dietary supplementation with RS4-type resistant starch attenuates high-fat diet-induced obesity more effectively than RS2 in C57BL/6J mice, which may be attributable to lower postprandial GIP and increased fat catabolism in the liver. Irinotecan 44-47 gastric inhibitory polypeptide Mus musculus 203-206 20235794-5 2010 This article provides an overview of recent progress in irinotecan pharmacogenetics and discusses the clinical significance of the UGT1A1 genotype/haplotype with regard to severe irinotecan toxicity. Irinotecan 179-189 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 131-137 20028383-1 2010 Although individuals carrying the UGT1A1 allele *28 have an increased risk of severe toxicities associated with irinotecan, no phase I study has been conducted based on the polymorphism. Irinotecan 112-122 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 34-40 20028383-12 2010 The recommended starting doses of biweekly irinotecan for phase II/III were 150 mg/m(2) for patients with the UGT1A1 6/6 genotype and 70 mg/m(2) for those with the 6/7 genotype, respectively. Irinotecan 43-53 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 110-116 19931604-0 2010 Relations between strain and gender dependencies of irinotecan toxicity and UGT1A1, CES2 and TOP1 expressions in mice. Irinotecan 52-62 UDP glucuronosyltransferase 1 family, polypeptide A1 Mus musculus 76-82 20235794-2 2010 Irinotecan is one of the models for personalized medicine based on pharmacogenetics, and a number of clinical studies have revealed significant associations between UGT1A1*28 and irinotecan toxicity. Irinotecan 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 165-171 20235794-2 2010 Irinotecan is one of the models for personalized medicine based on pharmacogenetics, and a number of clinical studies have revealed significant associations between UGT1A1*28 and irinotecan toxicity. Irinotecan 179-189 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 165-171 19931604-0 2010 Relations between strain and gender dependencies of irinotecan toxicity and UGT1A1, CES2 and TOP1 expressions in mice. Irinotecan 52-62 carboxylesterase 2C Mus musculus 84-88 19931604-2 2010 CPT-11 is bio-activated through CES, detoxified through UGT1A1 and inhibits TOP1. Irinotecan 0-6 UDP glucuronosyltransferase 1 family, polypeptide A1 Mus musculus 56-62 19931604-11 2010 Genetic background and gender significantly altered the molecular prediction of irinotecan toxicity by UGT1A1, CES2 and TOP1 mRNA expressions. Irinotecan 80-90 UDP glucuronosyltransferase 1 family, polypeptide A1 Mus musculus 103-109 19931604-11 2010 Genetic background and gender significantly altered the molecular prediction of irinotecan toxicity by UGT1A1, CES2 and TOP1 mRNA expressions. Irinotecan 80-90 carboxylesterase 2C Mus musculus 111-115 20038727-1 2010 PURPOSE We aimed to identify the maximum-tolerated dose (MTD) of irinotecan in patients with cancer with UGT1A1*1/*1 and *1/*28 genotypes. Irinotecan 65-75 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 105-111 19889793-7 2010 In addition to 4MUG, the PS(serosal) of the glucuronide conjugates of 7-ethyl-10-hydroxycamptothecin (SN-38) and acetaminophen in the jejunal everted sacs were also significantly reduced in Mrp3(-/-) mice compared with wild-type mice. Irinotecan 70-100 ATP-binding cassette, sub-family C (CFTR/MRP), member 3 Mus musculus 190-194 19652968-0 2010 Dynamic monitoring the TCR CDR3 spectratypes in patients with metastatic CRC treated with a combination of bevacizumab, irinotecan, fluorouracil, and leucovorin. Irinotecan 120-130 T cell receptor beta variable 20/OR9-2 (non-functional) Homo sapiens 23-26 19932091-2 2010 In addition, homozygous subjects for UGT1A128 genotype may suffer from severe irinotecan toxicity or jaundice during treatment with the protease inhibitor atazanavir. Irinotecan 78-88 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 37-42 19889793-7 2010 In addition to 4MUG, the PS(serosal) of the glucuronide conjugates of 7-ethyl-10-hydroxycamptothecin (SN-38) and acetaminophen in the jejunal everted sacs were also significantly reduced in Mrp3(-/-) mice compared with wild-type mice. Irinotecan 102-107 ATP-binding cassette, sub-family C (CFTR/MRP), member 3 Mus musculus 190-194 20068078-0 2010 A CYP3A4 phenotype-based dosing algorithm for individualized treatment of irinotecan. Irinotecan 74-84 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 2-8 20096102-1 2010 BACKGROUND: UDP-glucuronosyltransferase 1A1 (UGT1A1) is a pivotal enzyme involved in metabolism of SN-38, the active metabolite of irinotecan commonly used to treat metastatic colorectal cancer. Irinotecan 99-104 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-43 20096102-1 2010 BACKGROUND: UDP-glucuronosyltransferase 1A1 (UGT1A1) is a pivotal enzyme involved in metabolism of SN-38, the active metabolite of irinotecan commonly used to treat metastatic colorectal cancer. Irinotecan 99-104 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 45-51 20096102-1 2010 BACKGROUND: UDP-glucuronosyltransferase 1A1 (UGT1A1) is a pivotal enzyme involved in metabolism of SN-38, the active metabolite of irinotecan commonly used to treat metastatic colorectal cancer. Irinotecan 131-141 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-43 20096102-1 2010 BACKGROUND: UDP-glucuronosyltransferase 1A1 (UGT1A1) is a pivotal enzyme involved in metabolism of SN-38, the active metabolite of irinotecan commonly used to treat metastatic colorectal cancer. Irinotecan 131-141 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 45-51 20068078-1 2010 PURPOSE: Irinotecan, the prodrug of SN-38, is extensively metabolized by cytochrome P450-3A4 (CYP3A4). Irinotecan 9-19 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 73-92 20068078-1 2010 PURPOSE: Irinotecan, the prodrug of SN-38, is extensively metabolized by cytochrome P450-3A4 (CYP3A4). Irinotecan 9-19 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 94-100 20068078-1 2010 PURPOSE: Irinotecan, the prodrug of SN-38, is extensively metabolized by cytochrome P450-3A4 (CYP3A4). Irinotecan 36-41 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 73-92 20068078-1 2010 PURPOSE: Irinotecan, the prodrug of SN-38, is extensively metabolized by cytochrome P450-3A4 (CYP3A4). Irinotecan 36-41 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 94-100 20068078-2 2010 A randomized trial was done to assess the utility of an algorithm for individualized irinotecan dose calculation based on a priori CYP3A4 activity measurements by the midazolam clearance test. Irinotecan 85-95 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 131-137 20068078-11 2010 CONCLUSIONS: Incorporation of CYP3A4 phenotyping in dose calculation resulted in an improved predictability of the pharmacokinetic and toxicity profile of irinotecan, thereby lowering the incidence of severe neutropenia. Irinotecan 155-165 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 30-36 20068078-12 2010 In combination with UGT1A1*28 genotyping, CYP3A4 phenotype determination should be explored further as a strategy for the individualization of irinotecan treatment. Irinotecan 143-153 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 42-48 20551639-5 2010 Treatment of FaDu tumor with a therapeutic dose of irinotecan resulted in depression of xCT protein levels, leading to tumor growth retardation and downregulation of GSH with increased reactive oxygen species (ROS). Irinotecan 51-61 solute carrier family 7 member 11 Homo sapiens 88-91 19495753-9 2010 In mouse xenograft models, TP300 showed antitumor activity against both BCRP-positive and -negative xenografts, whereas CPT-11 was less active against BCRP-positive xenografts. Irinotecan 120-126 ATP binding cassette subfamily G member 2 (Junior blood group) Mus musculus 151-155 20551639-7 2010 CONCLUSION: Depression of xCT protein by irinotecan resulted in downregulation of GSH and increase in ROS, which could be the other possible mechanisms of DNA damage by irinotecan. Irinotecan 41-51 solute carrier family 7 member 11 Homo sapiens 26-29 20551639-7 2010 CONCLUSION: Depression of xCT protein by irinotecan resulted in downregulation of GSH and increase in ROS, which could be the other possible mechanisms of DNA damage by irinotecan. Irinotecan 169-179 solute carrier family 7 member 11 Homo sapiens 26-29 19835560-6 2010 Variant alleles of UGT1A1 are less capable of conjugating and eliminating SN-38, the active form of the topoisomerase inhibitor irinotecan, and defective alleles for NAT2 are responsible for the well-described acetylation polymorphism that leads to impaired clearance of isoniazid and other agents. Irinotecan 74-79 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 19-25 20205661-6 2010 Furthermore variations in the UGT1A gene and the ABCB1 gene influence irinotecan metabolism and disposition. Irinotecan 70-80 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 30-35 19835560-6 2010 Variant alleles of UGT1A1 are less capable of conjugating and eliminating SN-38, the active form of the topoisomerase inhibitor irinotecan, and defective alleles for NAT2 are responsible for the well-described acetylation polymorphism that leads to impaired clearance of isoniazid and other agents. Irinotecan 128-138 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 19-25 20205661-6 2010 Furthermore variations in the UGT1A gene and the ABCB1 gene influence irinotecan metabolism and disposition. Irinotecan 70-80 ATP binding cassette subfamily B member 1 Homo sapiens 49-54 20067946-3 2010 Cetuximab, a chimeric monoclonal antibody (mAb) against the epidermal growth factor receptor (EGFR) is FDA approved as a single agent, or in combination with irinotecan, in both irinotecan-naive and refractory patients, and has additional efficacy in combination with oxaliplatin. Irinotecan 178-188 epidermal growth factor receptor Homo sapiens 60-92 19819285-7 2010 Adding EPO in human lymphocyte cultures in vitro and in P388 leukemia bearing mice in vivo in the presence of CPT-11 decreased SCEs levels and increased PRIs and MIs were observed compared with cells treated either in vitro or in vivo with CPT-11 alone, which shows that EPO protected cells from the toxic action of CPT-11. Irinotecan 110-116 erythropoietin Homo sapiens 7-10 19819285-7 2010 Adding EPO in human lymphocyte cultures in vitro and in P388 leukemia bearing mice in vivo in the presence of CPT-11 decreased SCEs levels and increased PRIs and MIs were observed compared with cells treated either in vitro or in vivo with CPT-11 alone, which shows that EPO protected cells from the toxic action of CPT-11. Irinotecan 110-116 erythropoietin Mus musculus 271-274 19819285-7 2010 Adding EPO in human lymphocyte cultures in vitro and in P388 leukemia bearing mice in vivo in the presence of CPT-11 decreased SCEs levels and increased PRIs and MIs were observed compared with cells treated either in vitro or in vivo with CPT-11 alone, which shows that EPO protected cells from the toxic action of CPT-11. Irinotecan 240-246 erythropoietin Homo sapiens 7-10 19819285-7 2010 Adding EPO in human lymphocyte cultures in vitro and in P388 leukemia bearing mice in vivo in the presence of CPT-11 decreased SCEs levels and increased PRIs and MIs were observed compared with cells treated either in vitro or in vivo with CPT-11 alone, which shows that EPO protected cells from the toxic action of CPT-11. Irinotecan 240-246 erythropoietin Homo sapiens 7-10 19819285-8 2010 EPO"s protective action on human peripheral lymphocytes in vitro and P388 cells in vivo from the topoisomerase I inhibitor CPT-11, lead us to propose it as a geno- and cytoprotective agent. Irinotecan 123-129 erythropoietin Homo sapiens 0-3 20798767-8 2010 The iAUC for glucose and insulin were lower following ingestion of RS4 compared with the GLU and PWB trials. Irinotecan 67-70 insulin Homo sapiens 25-32 20798767-10 2010 These data illustrate, for the first time, that directly substituting standard starch with RS4(XL), while matched for available carbohydrates, attenuated postprandial glucose and insulin levels in humans. Irinotecan 91-94 insulin Homo sapiens 179-186 21486535-1 2010 PURPOSE: Functional polymorphisms of the UGT1A1 gene, particularly the UGT1A1*28 variant, are associated with the severity of the bone marrow suppression in patients with metastatic colorectal cancer receiving irinotecan. Irinotecan 210-220 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 41-47 21486535-1 2010 PURPOSE: Functional polymorphisms of the UGT1A1 gene, particularly the UGT1A1*28 variant, are associated with the severity of the bone marrow suppression in patients with metastatic colorectal cancer receiving irinotecan. Irinotecan 210-220 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 71-77 20053762-10 2010 Irinotecan and gemcitabine antitumor activity was enhanced by SAR-020106 in vivo with minimal toxicity. Irinotecan 0-10 sarcosine dehydrogenase Homo sapiens 62-65 20377135-0 2010 The UGT1A1*28 genotype and the toxicity of low-dose irinotecan in patients with advanced lung cancer. Irinotecan 52-62 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 20377135-1 2010 The association between the UGT1A1*28 genotype and the severe toxicity of low-dose irinotecan has been controversial, and few studies have examined this association in patients with lung cancer. Irinotecan 83-93 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 28-34 20377135-2 2010 The aim of this study was to assess the association between the UGT1A1*28 genotype and the severe toxicity of low-dose irinotecan in Japanese patients with lung cancer. Irinotecan 119-129 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 64-70 20377135-11 2010 Therefore, low-dose irinotecan could be administered without reducing starting dose in patients with UGT1A1*28 genotype. Irinotecan 20-30 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 101-107 20067946-3 2010 Cetuximab, a chimeric monoclonal antibody (mAb) against the epidermal growth factor receptor (EGFR) is FDA approved as a single agent, or in combination with irinotecan, in both irinotecan-naive and refractory patients, and has additional efficacy in combination with oxaliplatin. Irinotecan 178-188 epidermal growth factor receptor Homo sapiens 94-98 20930093-0 2010 Effects of mannose-binding lectin polymorphisms on irinotecan-induced febrile neutropenia. Irinotecan 51-61 mannose binding lectin 2 Homo sapiens 11-33 23074403-38 2010 CONCLUSIONS: KRAS status is predictive of outcomes in cetuximab and panitumumab monotherapy, and in cetuximab-irinotecan combination therapy. Irinotecan 110-120 KRAS proto-oncogene, GTPase Homo sapiens 13-17 20930093-4 2010 Here, we investigated the effects of MBL2 genotypes on irinotecan-induced febrile neutropenia in patients with solid tumors. Irinotecan 55-65 mannose binding lectin 2 Homo sapiens 37-41 20930093-5 2010 PATIENTS AND METHODS: Irinotecan-treated patients were genotyped for the MBL2 gene. Irinotecan 22-32 mannose binding lectin 2 Homo sapiens 73-77 23074403-32 2010 CETUXIMAB-IRINOTECAN COMBINATION THERAPY: There is low GRADE evidence that testing for KRAS may optimize survival benefits in patients without the KRAS mutation (KRAS wildtype, or KRAS WT) compared to those with the mutation. Irinotecan 10-20 KRAS proto-oncogene, GTPase Homo sapiens 87-91 23074403-33 2010 However, cetuximab-irinotecan combination treatments based on KRAS status discount any effect of cetuximab in possibly reversing resistance to irinotecan in patients with the mutation, as observed effects were lower than for patients without the mutation. Irinotecan 19-29 KRAS proto-oncogene, GTPase Homo sapiens 62-66 20054236-10 2009 Interestingly, under normoxia, Plk2 deficient tumor cells demonstrated increased apoptosis in response to various chemotherapeutic agents including CPT -11 but increased resistance to apoptotic death after CPT-11 treatment under hypoxia, and tumor xenografts comprised of these Plk2-deficient cells were resistant to CPT -11. Irinotecan 148-155 polo like kinase 2 Homo sapiens 31-35 19920821-1 2009 BACKGROUND: Both irinotecan (CPT-11) and S-1 are active against colorectal cancer; however, as S-1 is a prodrug of 5-fluorouracil (5-FU), 5-FU and its metabolites might inhibit the antitumour effect of CPT-11. Irinotecan 202-208 proteasome 26S subunit, non-ATPase 1 Homo sapiens 41-44 19920821-1 2009 BACKGROUND: Both irinotecan (CPT-11) and S-1 are active against colorectal cancer; however, as S-1 is a prodrug of 5-fluorouracil (5-FU), 5-FU and its metabolites might inhibit the antitumour effect of CPT-11. Irinotecan 202-208 proteasome 26S subunit, non-ATPase 1 Homo sapiens 95-98 19920821-1 2009 BACKGROUND: Both irinotecan (CPT-11) and S-1 are active against colorectal cancer; however, as S-1 is a prodrug of 5-fluorouracil (5-FU), 5-FU and its metabolites might inhibit the antitumour effect of CPT-11. Irinotecan 17-27 proteasome 26S subunit, non-ATPase 1 Homo sapiens 95-98 19920821-1 2009 BACKGROUND: Both irinotecan (CPT-11) and S-1 are active against colorectal cancer; however, as S-1 is a prodrug of 5-fluorouracil (5-FU), 5-FU and its metabolites might inhibit the antitumour effect of CPT-11. Irinotecan 29-35 proteasome 26S subunit, non-ATPase 1 Homo sapiens 95-98 20054236-10 2009 Interestingly, under normoxia, Plk2 deficient tumor cells demonstrated increased apoptosis in response to various chemotherapeutic agents including CPT -11 but increased resistance to apoptotic death after CPT-11 treatment under hypoxia, and tumor xenografts comprised of these Plk2-deficient cells were resistant to CPT -11. Irinotecan 206-212 polo like kinase 2 Homo sapiens 31-35 20054236-10 2009 Interestingly, under normoxia, Plk2 deficient tumor cells demonstrated increased apoptosis in response to various chemotherapeutic agents including CPT -11 but increased resistance to apoptotic death after CPT-11 treatment under hypoxia, and tumor xenografts comprised of these Plk2-deficient cells were resistant to CPT -11. Irinotecan 317-324 polo like kinase 2 Homo sapiens 31-35 19536776-0 2009 The dual EGFR/HER-2 tyrosine kinase inhibitor lapatinib sensitizes colon and gastric cancer cells to the irinotecan active metabolite SN-38. Irinotecan 105-115 epidermal growth factor receptor Mus musculus 9-13 19536776-0 2009 The dual EGFR/HER-2 tyrosine kinase inhibitor lapatinib sensitizes colon and gastric cancer cells to the irinotecan active metabolite SN-38. Irinotecan 105-115 erb-b2 receptor tyrosine kinase 2 Mus musculus 14-19 19536776-0 2009 The dual EGFR/HER-2 tyrosine kinase inhibitor lapatinib sensitizes colon and gastric cancer cells to the irinotecan active metabolite SN-38. Irinotecan 134-139 epidermal growth factor receptor Mus musculus 9-13 19536776-0 2009 The dual EGFR/HER-2 tyrosine kinase inhibitor lapatinib sensitizes colon and gastric cancer cells to the irinotecan active metabolite SN-38. Irinotecan 134-139 erb-b2 receptor tyrosine kinase 2 Mus musculus 14-19 19608554-0 2009 Prolonged neutropenia after irinotecan-based chemotherapy in a child with polymorphisms of UGT1A1 and SLCO1B1. Irinotecan 28-38 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 91-97 19608554-0 2009 Prolonged neutropenia after irinotecan-based chemotherapy in a child with polymorphisms of UGT1A1 and SLCO1B1. Irinotecan 28-38 solute carrier organic anion transporter family member 1B1 Homo sapiens 102-109 19608554-1 2009 Genetic polymorphisms of uridine diphosphate glucuronosyl transferase 1A1 (UGT1A1), and SLCO1B1 coding organic anion-transporter polypeptide 1B1, are independent risk factors known to increase irinotecan toxicity in adults. Irinotecan 193-203 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 25-73 19608554-1 2009 Genetic polymorphisms of uridine diphosphate glucuronosyl transferase 1A1 (UGT1A1), and SLCO1B1 coding organic anion-transporter polypeptide 1B1, are independent risk factors known to increase irinotecan toxicity in adults. Irinotecan 193-203 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 75-81 19608554-1 2009 Genetic polymorphisms of uridine diphosphate glucuronosyl transferase 1A1 (UGT1A1), and SLCO1B1 coding organic anion-transporter polypeptide 1B1, are independent risk factors known to increase irinotecan toxicity in adults. Irinotecan 193-203 solute carrier organic anion transporter family member 1B1 Homo sapiens 88-95 19608554-5 2009 To our knowledge, this is the first case report of combined genotyping of both UGT1A1 and SLCO1B1 in a child with severe irinotecan toxicity. Irinotecan 121-131 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 79-85 19608554-5 2009 To our knowledge, this is the first case report of combined genotyping of both UGT1A1 and SLCO1B1 in a child with severe irinotecan toxicity. Irinotecan 121-131 solute carrier organic anion transporter family member 1B1 Homo sapiens 90-97 19415281-2 2009 Exploratory analyses of the impact of variability in uridine diphosphate glucuronosyltransferase 1A (UGT1A) genes on irinotecan metabolism and toxicity were conducted. Irinotecan 117-127 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 53-99 19663821-5 2009 Both HI-6 and irinotecan inhibited ChE/AChE activity but showed different levels of ChE inhibition in plasma and AChE inhibition in the liver and brain tissue. Irinotecan 14-24 butyrylcholinesterase Homo sapiens 35-38 19663821-5 2009 Both HI-6 and irinotecan inhibited ChE/AChE activity but showed different levels of ChE inhibition in plasma and AChE inhibition in the liver and brain tissue. Irinotecan 14-24 acetylcholinesterase (Cartwright blood group) Homo sapiens 39-43 19663821-5 2009 Both HI-6 and irinotecan inhibited ChE/AChE activity but showed different levels of ChE inhibition in plasma and AChE inhibition in the liver and brain tissue. Irinotecan 14-24 butyrylcholinesterase Homo sapiens 40-43 19663821-5 2009 Both HI-6 and irinotecan inhibited ChE/AChE activity but showed different levels of ChE inhibition in plasma and AChE inhibition in the liver and brain tissue. Irinotecan 14-24 acetylcholinesterase (Cartwright blood group) Homo sapiens 113-117 19415281-2 2009 Exploratory analyses of the impact of variability in uridine diphosphate glucuronosyltransferase 1A (UGT1A) genes on irinotecan metabolism and toxicity were conducted. Irinotecan 117-127 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 101-106 19415281-8 2009 UGT1A1 polymorphisms were associated with variability in irinotecan PK. Irinotecan 57-67 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 19885570-7 2009 The combination of safingol/irinotecan at 1:1 molar ratio was found to be additive in HT-29 cells (CI=0.94) and synergistic in LS-174T cells (CI=0.68), and resulted in concentration- and time-dependent down-regulation of p-PKC and p-MARCKS. Irinotecan 28-38 myristoylated alanine rich protein kinase C substrate Homo sapiens 233-239 19909340-5 2009 When ABCG2 wild type-expressing cells were incubated with various N-linked glycosylation inhibitors, tunicamycin profoundly suppressed the protein expression level of ABCG2 and, accordingly, reduced the ABCG2-mediated cellular resistance to the cancer chemotherapeutic SN-38. Irinotecan 269-274 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 5-10 19909340-5 2009 When ABCG2 wild type-expressing cells were incubated with various N-linked glycosylation inhibitors, tunicamycin profoundly suppressed the protein expression level of ABCG2 and, accordingly, reduced the ABCG2-mediated cellular resistance to the cancer chemotherapeutic SN-38. Irinotecan 269-274 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 167-172 19909340-5 2009 When ABCG2 wild type-expressing cells were incubated with various N-linked glycosylation inhibitors, tunicamycin profoundly suppressed the protein expression level of ABCG2 and, accordingly, reduced the ABCG2-mediated cellular resistance to the cancer chemotherapeutic SN-38. Irinotecan 269-274 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 167-172 19883083-4 2009 Tdp1 inhibitors have been regarded as potential therapeutics in combination with Top1 inhibitors, such as the camptothecin derivatives, topotecan, and irinotecan, which are used to treat human cancers. Irinotecan 151-161 tyrosyl-DNA phosphodiesterase 1 Homo sapiens 0-4 20093746-12 2009 rhTGF-beta1 and/or CPT-11 may potentate resistance to chemotherapy by increasing HSP and MRP expression but, on the other hand, they may limit tumour cell spread by decreasing the level of some soluble mediators of inflammation (IL-6 and NO). Irinotecan 19-25 heat shock protein 90 beta family member 2, pseudogene Homo sapiens 81-84 20093746-12 2009 rhTGF-beta1 and/or CPT-11 may potentate resistance to chemotherapy by increasing HSP and MRP expression but, on the other hand, they may limit tumour cell spread by decreasing the level of some soluble mediators of inflammation (IL-6 and NO). Irinotecan 19-25 ATP binding cassette subfamily C member 1 Homo sapiens 89-92 20093746-12 2009 rhTGF-beta1 and/or CPT-11 may potentate resistance to chemotherapy by increasing HSP and MRP expression but, on the other hand, they may limit tumour cell spread by decreasing the level of some soluble mediators of inflammation (IL-6 and NO). Irinotecan 19-25 interleukin 6 Homo sapiens 229-233 20071295-2 2009 Its active metabolite, 7-ethyl-10 hydroxycamptothecin (SN-38) is glucuronidated by a uridine-diphosphoglucuronosyltransferase (UGT1A1) to form an inactive metabolite. Irinotecan 23-53 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 127-133 20071295-2 2009 Its active metabolite, 7-ethyl-10 hydroxycamptothecin (SN-38) is glucuronidated by a uridine-diphosphoglucuronosyltransferase (UGT1A1) to form an inactive metabolite. Irinotecan 55-60 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 127-133 19915942-3 2009 The immoglobulin (Ig) G1 EGFR-targeting monoclonal antibody (mAb), cetuximab, has been shown to provide significant clinical benefits when added to standard irinotecan- and oxaliplatin-containing chemotherapy regimens, first-line, in patients with KRAS wild-type mCRC. Irinotecan 157-167 epidermal growth factor receptor Homo sapiens 25-29 19770637-0 2009 UGT1A1 genotyping: a predictor of irinotecan-associated side effects and drug efficacy? Irinotecan 34-44 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 19858398-10 2009 Trends (of doubtful significance) were seen for associations of XRCC1, ERCC2, and GSTP1 with toxicity during irinotecan regimens: XRCC1, primary end point, any irinotecan-containing regimen (P = .045); ERCC2, secondary end point, irinotecan alone (P = .003); GSTP1, secondary end point; IrFU (P = .039); and irinotecan alone (P = .05). Irinotecan 109-119 glutathione S-transferase pi 1 Homo sapiens 82-87 19858398-13 2009 Further investigation of XRCC1, ERCC2, and GSTP1 as potential predictors of irinotecan toxicity is warranted. Irinotecan 76-86 glutathione S-transferase pi 1 Homo sapiens 43-48 19770637-3 2009 SN-38 is inactivated by uridine disphosphate glucuronosyl transferase 1 (UGT1A1) into the inactive compound SN-38G, which is excreted with the bile.This review concentrates on a critical evaluation of UGT1A1 gene polymorphism as a predictor of toxicity and treatment efficacy in patients who received irinotecan for metastatic colorectal cancer. Irinotecan 0-5 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 73-79 19770637-3 2009 SN-38 is inactivated by uridine disphosphate glucuronosyl transferase 1 (UGT1A1) into the inactive compound SN-38G, which is excreted with the bile.This review concentrates on a critical evaluation of UGT1A1 gene polymorphism as a predictor of toxicity and treatment efficacy in patients who received irinotecan for metastatic colorectal cancer. Irinotecan 0-5 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 201-207 19770637-3 2009 SN-38 is inactivated by uridine disphosphate glucuronosyl transferase 1 (UGT1A1) into the inactive compound SN-38G, which is excreted with the bile.This review concentrates on a critical evaluation of UGT1A1 gene polymorphism as a predictor of toxicity and treatment efficacy in patients who received irinotecan for metastatic colorectal cancer. Irinotecan 301-311 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 73-79 19673886-0 2009 Prolonged low-dose administration of the cyclooxygenase-2 inhibitor celecoxib enhances the antitumor activity of irinotecan against neuroblastoma xenografts. Irinotecan 113-123 prostaglandin-endoperoxide synthase 2 Homo sapiens 41-57 19796956-3 2009 Results have also shown that 2,9-bis(3-(dimethylamino)propoxy)-6-(4-(3-(dimethylamino)propoxy)phenyl)-11H-indeno[1,2-c]quinolin-11-one (17), which exhibited GI(50) of 0.60 and 0.68 microM against the growth of Hep G2 and A549, respectively, was more active than the positive topotecan and irinotecan. Irinotecan 289-299 DNL-type zinc finger Homo sapiens 210-213 19673886-6 2009 Induction of apoptosis by CPT-11 with and without celecoxib was associated with the up-regulation of Bax expression and the down-regulation of Bcl-2 expression. Irinotecan 26-32 BCL2 associated X, apoptosis regulator Homo sapiens 101-104 19673886-6 2009 Induction of apoptosis by CPT-11 with and without celecoxib was associated with the up-regulation of Bax expression and the down-regulation of Bcl-2 expression. Irinotecan 26-32 BCL2 apoptosis regulator Homo sapiens 143-148 20037407-10 2009 In spite of combined S-1 and CPT-11 chemotherapy, the CEA level increased, and lymph nodes were getting larger. Irinotecan 29-35 CEA cell adhesion molecule 3 Homo sapiens 54-57 19917537-1 2009 Therapy for metastatic colorectal cancer has been improved in terms of response rate, time to progression and overall survival by the emergence of anti-EGFR monoclonal antibodies (cetuximab and panitumumab) in combination with standard cytotoxic chemotherapy (oxaliplatin or CPT-11-based combinations). Irinotecan 275-281 epidermal growth factor receptor Homo sapiens 152-156 19738126-1 2009 PURPOSE: To study the power of the epidermal growth factor receptor (EGFR) epiregulin (EREG) and amphiregulin (AREG) ligands" expression in primary tumors to predict the outcome in patients with chemorefractory metastatic colorectal cancer (cmCRC) treated with the combination of cetuximab and irinotecan. Irinotecan 294-304 epidermal growth factor receptor Homo sapiens 35-67 19859999-0 2009 UGT1A1 gene polymorphism: impact on toxicity and efficacy of irinotecan-based regimens in metastatic colorectal cancer. Irinotecan 61-71 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 19859999-1 2009 AIM: To investigate the correlation between uridine diphosphate glucuronosyl transferase 1A1 (UGT1A1) gene polymorphisms and irinotecan-associated side effects and parameters of drug efficacy in patients with metastatic colorectal cancer (mCRC) receiving a low-dose weekly irinotecan chemotherapeutic regimen. Irinotecan 125-135 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 44-92 19859999-1 2009 AIM: To investigate the correlation between uridine diphosphate glucuronosyl transferase 1A1 (UGT1A1) gene polymorphisms and irinotecan-associated side effects and parameters of drug efficacy in patients with metastatic colorectal cancer (mCRC) receiving a low-dose weekly irinotecan chemotherapeutic regimen. Irinotecan 125-135 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 94-100 19859999-2 2009 METHODS: Genotypes were retrospectively evaluated by gene scan analysis on the ABI 310 sequencer of the TATAA box in the promoter region of the UGT1A1 gene in blood samples from 105 patients who had received 1st line irinotecan-based chemotherapy for mCRC. Irinotecan 217-227 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 144-150 19859999-7 2009 CONCLUSION: This analysis demonstrates the non-significant influence of the UGT1A1 gene polymorphism on efficacy and rate of irinotecan-associated toxicity in mCRC patients receiving low-dose irinotecan based chemotherapy. Irinotecan 125-135 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 76-82 19738126-1 2009 PURPOSE: To study the power of the epidermal growth factor receptor (EGFR) epiregulin (EREG) and amphiregulin (AREG) ligands" expression in primary tumors to predict the outcome in patients with chemorefractory metastatic colorectal cancer (cmCRC) treated with the combination of cetuximab and irinotecan. Irinotecan 294-304 epidermal growth factor receptor Homo sapiens 69-73 19738126-13 2009 CONCLUSION: Expression of EGFR ligands in primary tumors significantly predicts outcome in KRAS WT cmCRC treated with cetuximab and irinotecan. Irinotecan 132-142 epidermal growth factor receptor Homo sapiens 26-30 19738126-13 2009 CONCLUSION: Expression of EGFR ligands in primary tumors significantly predicts outcome in KRAS WT cmCRC treated with cetuximab and irinotecan. Irinotecan 132-142 KRAS proto-oncogene, GTPase Homo sapiens 91-95 19765103-0 2009 Irinotecan-induced mucositis manifesting as diarrhoea corresponds with an amended intestinal flora and mucin profile. Irinotecan 0-10 solute carrier family 13 member 2 Rattus norvegicus 103-108 19765103-3 2009 Bacterial beta-glucuronidase is thought to be involved in the metabolism of irinotecan, implicating the intestinal flora. Irinotecan 76-86 glucuronidase, beta Rattus norvegicus 10-28 19765103-12 2009 In conclusion, irinotecan-induced diarrhoea may be caused by an increase in some beta-glucuronidase-producing bacteria, especially E. coli, exacerbating the toxicity of active metabolites. Irinotecan 15-25 glucuronidase, beta Rattus norvegicus 81-99 19696792-0 2009 Single nucleotide polymorphism in ABCG2 is associated with irinotecan-induced severe myelosuppression. Irinotecan 59-69 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 34-39 19696792-6 2009 Although only limited subjects were investigated, our results suggested that a genetic polymorphism in ABCG2 might alter the transport activity for the drug and elevate the systemic circulation level of irinotecan, leading to severe myelosuppression. Irinotecan 203-213 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 103-108 19332770-1 2009 Bevacizumab is a humanized monoclonal antibody against vascular endothelial growth factor (VEGF) that has efficacy in recurrent malignant gliomas, particularly in combination with irinotecan. Irinotecan 180-190 vascular endothelial growth factor A Homo sapiens 55-89 19789330-0 2009 CEACAM5-targeted therapy of human colonic and pancreatic cancer xenografts with potent labetuzumab-SN-38 immunoconjugates. Irinotecan 99-104 CEA cell adhesion molecule 5 Homo sapiens 0-7 19789330-1 2009 PURPOSE: To improve the efficacy and reduce the gastrointestinal toxicity of the cancer prodrug, CPT-11, we have developed immunoconjugates of its active form, SN-38, and an anti-CEACAM5 antibody for targeted chemotherapy. Irinotecan 97-103 CEA cell adhesion molecule 5 Homo sapiens 179-186 19789330-2 2009 EXPERIMENTAL DESIGN: SN-38 conjugates of the anti-CEACAM5 monoclonal antibody, labetuzumab (hMN-14), varying in the nature of the cross-linker attachment at the drug"s 20-hydroxyl position, were evaluated in vitro, in metastatic and/or s.c. human colonic and pancreatic cancer xenografts in nude mice using appropriate controls, and in a CEACAM5-negative tumor model. Irinotecan 21-26 CEA cell adhesion molecule 5 Homo sapiens 50-57 20021826-8 2009 Under hypoxic conditions, the expression of HIF-1alpha and VEGF increased as time accumulated, Bevacizumab combined with SN-38 almost completely inhibited the expression of HIF-1alpha and VEGF. Irinotecan 121-126 hypoxia inducible factor 1 subunit alpha Homo sapiens 44-54 20021826-8 2009 Under hypoxic conditions, the expression of HIF-1alpha and VEGF increased as time accumulated, Bevacizumab combined with SN-38 almost completely inhibited the expression of HIF-1alpha and VEGF. Irinotecan 121-126 hypoxia inducible factor 1 subunit alpha Homo sapiens 173-183 20021826-8 2009 Under hypoxic conditions, the expression of HIF-1alpha and VEGF increased as time accumulated, Bevacizumab combined with SN-38 almost completely inhibited the expression of HIF-1alpha and VEGF. Irinotecan 121-126 vascular endothelial growth factor A Homo sapiens 188-192 20021826-10 2009 CONCLUSION: These findings suggest the anti-proliferation effect of bevacizumab and SN-38 was schedule-dependent, and the synergistic effect of Bevacizumab and SN-38 was related to drug modulation of the HIF-1alpha and MAP kinase pathway. Irinotecan 160-165 hypoxia inducible factor 1 subunit alpha Homo sapiens 204-214 19332770-1 2009 Bevacizumab is a humanized monoclonal antibody against vascular endothelial growth factor (VEGF) that has efficacy in recurrent malignant gliomas, particularly in combination with irinotecan. Irinotecan 180-190 vascular endothelial growth factor A Homo sapiens 91-95 19785749-1 2009 BACKGROUND: The anti-VEGF antibody bevacizumab associated with an irinotecan or oxaliplatin-based chemotherapy was proved to be superior to the chemotherapy alone in first or second line treatment of metastatic colorectal cancer (mCRC). Irinotecan 66-76 vascular endothelial growth factor A Homo sapiens 21-25 19644023-4 2009 Irinotecan toxicity in colorectal cancer is more common in patients with reduced-activity UGT1A alleles, resulting in excessive exposure to the potent SN-38 metabolite. Irinotecan 0-10 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 90-95 19687340-0 2009 Tyrosine kinase inhibitor enhances the bioavailability of oral irinotecan in pediatric patients with refractory solid tumors. Irinotecan 63-73 TXK tyrosine kinase Homo sapiens 0-15 19728911-12 2009 CONCLUSIONS: Transient CEA surges can be observed in Chinese MCRC patients receiving oxaplatin or irinotecan-based chemotherapy, which does not indicate tumor progression, but good therapeutic efficacy. Irinotecan 98-108 CEA cell adhesion molecule 3 Homo sapiens 23-26 19517472-1 2009 BACKGROUND: The objective of this study was to examine the cost effectiveness of using a pharmacogenetic test for uridine diphosphate glycosyltransferase 1A1*28 (UGT1A1*28) variant homozygosity before administering irinotecan to patients with metastatic colorectal cancer. Irinotecan 215-225 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 162-168 19517472-4 2009 With genetic testing, irinotecan dosage was reduced 25% in homozygotes with the UGT1A1*28 variant allele. Irinotecan 22-32 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 80-86 19755821-1 2009 SN-38, an active metabolite of irinotecan (CPT-11), is glucuronidated into SN-38G mainly by UGT1A1, during detoxification. Irinotecan 0-5 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 92-98 19755821-1 2009 SN-38, an active metabolite of irinotecan (CPT-11), is glucuronidated into SN-38G mainly by UGT1A1, during detoxification. Irinotecan 31-41 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 92-98 19755821-1 2009 SN-38, an active metabolite of irinotecan (CPT-11), is glucuronidated into SN-38G mainly by UGT1A1, during detoxification. Irinotecan 43-49 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 92-98 19755821-1 2009 SN-38, an active metabolite of irinotecan (CPT-11), is glucuronidated into SN-38G mainly by UGT1A1, during detoxification. Irinotecan 75-80 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 92-98 19755821-2 2009 However, significant interindividual pharmacokinetic variability in SN-38 is caused by factors, including inherited predispositions that may affect the function and expression of UGT1A1. Irinotecan 68-73 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 179-185 20084161-8 2009 The US Food and Drug Administration has approved the UDP-glucuronosyltransferase 1A1 test in patients treated with irinotecan, and additional approval of newer tests is anticipated. Irinotecan 115-125 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 53-84 19493273-4 2009 As with gefitinib, erlotinib effectively reversed BCRP-mediated resistance to SN-38 (7-ethyl-10-hydroxycamptothecin) and mitoxantrone. Irinotecan 78-83 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 50-54 19493273-4 2009 As with gefitinib, erlotinib effectively reversed BCRP-mediated resistance to SN-38 (7-ethyl-10-hydroxycamptothecin) and mitoxantrone. Irinotecan 85-115 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 50-54 19644023-4 2009 Irinotecan toxicity in colorectal cancer is more common in patients with reduced-activity UGT1A alleles, resulting in excessive exposure to the potent SN-38 metabolite. Irinotecan 151-156 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 90-95 19712476-0 2009 Epidermal Growth Factor Receptor (EGFR) gene copy number (GCN) correlates with clinical activity of irinotecan-cetuximab in K-RAS wild-type colorectal cancer: a fluorescence in situ (FISH) and chromogenic in situ hybridization (CISH) analysis. Irinotecan 100-110 KRAS proto-oncogene, GTPase Homo sapiens 124-129 19603018-0 2009 KRAS codon 61, 146 and BRAF mutations predict resistance to cetuximab plus irinotecan in KRAS codon 12 and 13 wild-type metastatic colorectal cancer. Irinotecan 75-85 KRAS proto-oncogene, GTPase Homo sapiens 0-4 20719167-2 2009 Irinotecan, oxidized by CYP3A4 to produce inactive compounds, is used for treatment of various cancers including advanced non small cell lung cancer (NSCLC) patients. Irinotecan 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 24-30 20719167-3 2009 CYP3A4(*)16B polymorphism was associated with decreased metabolism of irrinotecan. Irinotecan 70-81 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 0-6 20719167-4 2009 Irinotecan is also metabolized by carboxylesterase to its principal active metabolite, SN-38, which is subsequently glucuronidated by UGT1As to form the inactive compound SN-38G. Irinotecan 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 134-138 20719167-4 2009 Irinotecan is also metabolized by carboxylesterase to its principal active metabolite, SN-38, which is subsequently glucuronidated by UGT1As to form the inactive compound SN-38G. Irinotecan 87-92 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 134-138 20719167-4 2009 Irinotecan is also metabolized by carboxylesterase to its principal active metabolite, SN-38, which is subsequently glucuronidated by UGT1As to form the inactive compound SN-38G. Irinotecan 171-176 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 134-138 20719167-5 2009 UGT1A1(*)28 and UGT1A1(*)6 polymorphisms were useful for predicting severe toxicity with NSCLC patients treated with irinotecan-based chemotherapy. Irinotecan 117-127 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 20719167-5 2009 UGT1A1(*)28 and UGT1A1(*)6 polymorphisms were useful for predicting severe toxicity with NSCLC patients treated with irinotecan-based chemotherapy. Irinotecan 117-127 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 16-22 19603018-0 2009 KRAS codon 61, 146 and BRAF mutations predict resistance to cetuximab plus irinotecan in KRAS codon 12 and 13 wild-type metastatic colorectal cancer. Irinotecan 75-85 B-Raf proto-oncogene, serine/threonine kinase Homo sapiens 23-27 19603018-0 2009 KRAS codon 61, 146 and BRAF mutations predict resistance to cetuximab plus irinotecan in KRAS codon 12 and 13 wild-type metastatic colorectal cancer. Irinotecan 75-85 KRAS proto-oncogene, GTPase Homo sapiens 89-93 19433978-17 2009 Our case is the first clinical piece of evidence that demonstrates an increased sensitivity to camptothecin-11 and a reduced topoisomerase I relaxation activity in BRCA2 associated pancreatic cancer. Irinotecan 95-110 BRCA2 DNA repair associated Homo sapiens 164-169 19664991-5 2009 Furthermore, we found that specifically inhibiting human DLL4 in the tumor, either alone or in combination with the chemotherapeutic agent irinotecan, reduced cancer stem cell frequency, as shown by flow cytometric and in vivo tumorigenicity studies. Irinotecan 139-149 delta like canonical Notch ligand 4 Homo sapiens 57-61 19491654-7 2009 In SK-OV-3 cells, but not in OVCAR-3 cells, CPT-11/SN-38 exposures alone increased p21(waf1/cip1) expression. Irinotecan 44-50 cyclin dependent kinase inhibitor 1A Homo sapiens 83-96 19653336-6 2009 Irinotecan sensitizes p53 wild type, mutant and null cells to Fas-mediated cell apoptosis in CRC cells. Irinotecan 0-10 tumor protein p53 Homo sapiens 22-25 19653336-7 2009 Wild type p53 cells were more sensitive to irinotecan than mutated p53. Irinotecan 43-53 tumor protein p53 Homo sapiens 10-13 19653336-8 2009 Irinotecan has an anti-VEGF effect inhibiting endothelial cell proliferation, increasing apoptosis and reducing microvascular density which is only limited by irinotecan toxicity levels. Irinotecan 0-10 vascular endothelial growth factor A Homo sapiens 23-27 19653336-10 2009 p53 and VEGF status of the patients" tumour is likely to affect the responsiveness of CRC to irinotecan. Irinotecan 93-103 tumor protein p53 Homo sapiens 0-3 19653336-10 2009 p53 and VEGF status of the patients" tumour is likely to affect the responsiveness of CRC to irinotecan. Irinotecan 93-103 vascular endothelial growth factor A Homo sapiens 8-12 19491654-5 2009 In OVCAR-3 cells, p53 gene therapy inhibited the cell growth and sensitized cells to CPT-11/SN-38, but not to docetaxel. Irinotecan 85-91 tumor protein p53 Homo sapiens 18-21 19491654-7 2009 In SK-OV-3 cells, but not in OVCAR-3 cells, CPT-11/SN-38 exposures alone increased p21(waf1/cip1) expression. Irinotecan 51-56 cyclin dependent kinase inhibitor 1A Homo sapiens 83-96 19491654-5 2009 In OVCAR-3 cells, p53 gene therapy inhibited the cell growth and sensitized cells to CPT-11/SN-38, but not to docetaxel. Irinotecan 92-97 tumor protein p53 Homo sapiens 18-21 19457654-6 2009 In conclusion, in this largest study on capecitabine with or without irinotecan to date we found a predictive value of DPD expression. Irinotecan 69-79 dihydropyrimidine dehydrogenase Homo sapiens 119-122 19359405-2 2009 For example, irinotecan, a carbamate prodrug used in the treatment of colorectal cancer, is biotransformed in vivo by CES2 in intestine and liver, thereby producing a potent topoisomerase I inhibitor. Irinotecan 13-23 carboxylesterase 2H Mus musculus 118-122 19491654-11 2009 These results support the combination of p53 gene therapy with topoisomerase I inhibitors SN-38/CPT-11 when tumour cells contain mutated p53. Irinotecan 90-95 tumor protein p53 Homo sapiens 137-140 19491654-11 2009 These results support the combination of p53 gene therapy with topoisomerase I inhibitors SN-38/CPT-11 when tumour cells contain mutated p53. Irinotecan 96-102 tumor protein p53 Homo sapiens 137-140 19622774-5 2009 Depletion of Drg1 by small interfering RNA induced up-regulation of Bim and its accumulation in the mitochondria, which correlated with loss of mitochondrial membrane potential and induction of apoptosis in cells exposed to SN-38. Irinotecan 224-229 developmentally regulated GTP binding protein 1 Homo sapiens 13-17 19622774-5 2009 Depletion of Drg1 by small interfering RNA induced up-regulation of Bim and its accumulation in the mitochondria, which correlated with loss of mitochondrial membrane potential and induction of apoptosis in cells exposed to SN-38. Irinotecan 224-229 BCL2 like 11 Homo sapiens 68-71 19436196-2 2009 We hypothesized that the expression of topoisomerase I (Topo I) and thymidylate synthase (TS) may help predict the treatment response in patients undergoing irinotecan and capecitabine-based chemoradiation. Irinotecan 157-167 thymidylate synthetase Homo sapiens 68-88 19436196-2 2009 We hypothesized that the expression of topoisomerase I (Topo I) and thymidylate synthase (TS) may help predict the treatment response in patients undergoing irinotecan and capecitabine-based chemoradiation. Irinotecan 157-167 thymidylate synthetase Homo sapiens 90-92 19620803-1 2009 UNLABELLED: Irinotecan hydrochloride (CPT-11) is converted to SN-38 by carboxylesterase, SN-38 is conjugated by UDP-glucuronosyl- transferase (UGT) to SN-38G. Irinotecan 62-67 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 112-141 19620803-1 2009 UNLABELLED: Irinotecan hydrochloride (CPT-11) is converted to SN-38 by carboxylesterase, SN-38 is conjugated by UDP-glucuronosyl- transferase (UGT) to SN-38G. Irinotecan 38-44 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 143-146 19620803-1 2009 UNLABELLED: Irinotecan hydrochloride (CPT-11) is converted to SN-38 by carboxylesterase, SN-38 is conjugated by UDP-glucuronosyl- transferase (UGT) to SN-38G. Irinotecan 62-67 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 143-146 19620803-1 2009 UNLABELLED: Irinotecan hydrochloride (CPT-11) is converted to SN-38 by carboxylesterase, SN-38 is conjugated by UDP-glucuronosyl- transferase (UGT) to SN-38G. Irinotecan 12-36 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 112-141 19620803-1 2009 UNLABELLED: Irinotecan hydrochloride (CPT-11) is converted to SN-38 by carboxylesterase, SN-38 is conjugated by UDP-glucuronosyl- transferase (UGT) to SN-38G. Irinotecan 89-94 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 112-141 19620803-1 2009 UNLABELLED: Irinotecan hydrochloride (CPT-11) is converted to SN-38 by carboxylesterase, SN-38 is conjugated by UDP-glucuronosyl- transferase (UGT) to SN-38G. Irinotecan 12-36 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 143-146 19620803-1 2009 UNLABELLED: Irinotecan hydrochloride (CPT-11) is converted to SN-38 by carboxylesterase, SN-38 is conjugated by UDP-glucuronosyl- transferase (UGT) to SN-38G. Irinotecan 38-44 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 112-141 19620803-1 2009 UNLABELLED: Irinotecan hydrochloride (CPT-11) is converted to SN-38 by carboxylesterase, SN-38 is conjugated by UDP-glucuronosyl- transferase (UGT) to SN-38G. Irinotecan 89-94 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 143-146 19620803-1 2009 UNLABELLED: Irinotecan hydrochloride (CPT-11) is converted to SN-38 by carboxylesterase, SN-38 is conjugated by UDP-glucuronosyl- transferase (UGT) to SN-38G. Irinotecan 89-94 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 112-141 19620803-1 2009 UNLABELLED: Irinotecan hydrochloride (CPT-11) is converted to SN-38 by carboxylesterase, SN-38 is conjugated by UDP-glucuronosyl- transferase (UGT) to SN-38G. Irinotecan 89-94 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 143-146 19620808-1 2009 Severe neutropenia attributable to irinotecan hydrochloride (CPT-11) is reportedly associated with gene polymorphism of UGT1A1 related to its metabolism. Irinotecan 35-59 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 120-126 19620808-1 2009 Severe neutropenia attributable to irinotecan hydrochloride (CPT-11) is reportedly associated with gene polymorphism of UGT1A1 related to its metabolism. Irinotecan 61-67 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 120-126 19620808-9 2009 Nine days following administration of the first course, we found neutropenia of grade 3 and a fever of 38 degrees C. Analysis of the UGT1A1 gene polymorphism after symptom improvement revealed UGT1A1(*)28 homozygosity in both cases, which suggests that when FOLFIRI is conducted on a patient with homozygous UGT1A1(*)28, it is necessary to pay attention to neutropenia even with a CPT-11 dosage of 120 mg/m(2). Irinotecan 381-387 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 133-139 19620808-9 2009 Nine days following administration of the first course, we found neutropenia of grade 3 and a fever of 38 degrees C. Analysis of the UGT1A1 gene polymorphism after symptom improvement revealed UGT1A1(*)28 homozygosity in both cases, which suggests that when FOLFIRI is conducted on a patient with homozygous UGT1A1(*)28, it is necessary to pay attention to neutropenia even with a CPT-11 dosage of 120 mg/m(2). Irinotecan 381-387 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 193-199 19620808-9 2009 Nine days following administration of the first course, we found neutropenia of grade 3 and a fever of 38 degrees C. Analysis of the UGT1A1 gene polymorphism after symptom improvement revealed UGT1A1(*)28 homozygosity in both cases, which suggests that when FOLFIRI is conducted on a patient with homozygous UGT1A1(*)28, it is necessary to pay attention to neutropenia even with a CPT-11 dosage of 120 mg/m(2). Irinotecan 381-387 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 193-199 19531575-1 2009 Irinotecan is a topoisomerase-I (Top-I) inhibitor used for the treatment of colorectal cancer. Irinotecan 0-10 DNA topoisomerase I Homo sapiens 16-31 19531575-13 2009 If confirmed in other models, these results suggest that p16 and p53 status affects the 5-aza-irinotecan interaction. Irinotecan 94-104 cyclin dependent kinase inhibitor 2A Homo sapiens 57-60 19513514-14 2009 However, significant genotypic variation of UGT1A1, which was assumed to affect irinotecan toxicity, was not observed to affect RR, toxicity or survival. Irinotecan 80-90 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 44-50 19604089-1 2009 AIMS: A recent study found that variation in camptothecin pharmacodynamic genes (TOP1, PARP1, TDP1 and XRCC1) correlated with efficacy and risk of neutropenia in irinotecan-treated cancer patients (median dose: 180 mg/m2), which suggests that these genes might predict outcomes to irinotecan-based therapies. Irinotecan 162-172 poly(ADP-ribose) polymerase 1 Homo sapiens 87-92 19604089-1 2009 AIMS: A recent study found that variation in camptothecin pharmacodynamic genes (TOP1, PARP1, TDP1 and XRCC1) correlated with efficacy and risk of neutropenia in irinotecan-treated cancer patients (median dose: 180 mg/m2), which suggests that these genes might predict outcomes to irinotecan-based therapies. Irinotecan 162-172 tyrosyl-DNA phosphodiesterase 1 Homo sapiens 94-98 19604089-1 2009 AIMS: A recent study found that variation in camptothecin pharmacodynamic genes (TOP1, PARP1, TDP1 and XRCC1) correlated with efficacy and risk of neutropenia in irinotecan-treated cancer patients (median dose: 180 mg/m2), which suggests that these genes might predict outcomes to irinotecan-based therapies. Irinotecan 162-172 X-ray repair cross complementing 1 Homo sapiens 103-108 19604089-1 2009 AIMS: A recent study found that variation in camptothecin pharmacodynamic genes (TOP1, PARP1, TDP1 and XRCC1) correlated with efficacy and risk of neutropenia in irinotecan-treated cancer patients (median dose: 180 mg/m2), which suggests that these genes might predict outcomes to irinotecan-based therapies. Irinotecan 281-291 X-ray repair cross complementing 1 Homo sapiens 103-108 19531575-13 2009 If confirmed in other models, these results suggest that p16 and p53 status affects the 5-aza-irinotecan interaction. Irinotecan 94-104 tumor protein p53 Homo sapiens 65-68 19531575-1 2009 Irinotecan is a topoisomerase-I (Top-I) inhibitor used for the treatment of colorectal cancer. Irinotecan 0-10 DNA topoisomerase I Homo sapiens 33-38 19531575-3 2009 5-Aza may enhance irinotecan cytotoxicity by at least one of the following mechanisms: (a) Top-I promoter demethylation, (b) activation of genes involved in Top-I transcriptional regulation (p16 or Sp1), and (c) modulation of the cell cycle and apoptosis after DNA damage. Irinotecan 18-28 DNA topoisomerase I Homo sapiens 91-96 19531575-3 2009 5-Aza may enhance irinotecan cytotoxicity by at least one of the following mechanisms: (a) Top-I promoter demethylation, (b) activation of genes involved in Top-I transcriptional regulation (p16 or Sp1), and (c) modulation of the cell cycle and apoptosis after DNA damage. Irinotecan 18-28 DNA topoisomerase I Homo sapiens 157-162 19531575-3 2009 5-Aza may enhance irinotecan cytotoxicity by at least one of the following mechanisms: (a) Top-I promoter demethylation, (b) activation of genes involved in Top-I transcriptional regulation (p16 or Sp1), and (c) modulation of the cell cycle and apoptosis after DNA damage. Irinotecan 18-28 cyclin dependent kinase inhibitor 2A Homo sapiens 191-194 19531575-10 2009 p16 demethylation was also associated with enhanced cell cycle arrest after irinotecan treatment. Irinotecan 76-86 cyclin dependent kinase inhibitor 2A Homo sapiens 0-3 19217207-3 2009 Accordingly, TRAP1 levels were increased in HT-29 colorectal carcinoma cells resistant to 5-fluorouracil, oxaliplatin and irinotecan. Irinotecan 122-132 TNF receptor associated protein 1 Homo sapiens 13-18 19217207-5 2009 Interestingly, TRAP1 overexpression leads to 5-fluorouracil-, oxaliplatin- and irinotecan-resistant phenotypes in different neoplastic cells. Irinotecan 79-89 TNF receptor associated protein 1 Homo sapiens 15-20 19217207-6 2009 Conversely, the inhibition of TRAP1 activity by TRAP1 ATPase antagonist, shepherdin, increased the sensitivity to oxaliplatin and irinotecan in colorectal carcinoma cells resistant to the single agents. Irinotecan 130-140 TNF receptor associated protein 1 Homo sapiens 30-35 19217207-6 2009 Conversely, the inhibition of TRAP1 activity by TRAP1 ATPase antagonist, shepherdin, increased the sensitivity to oxaliplatin and irinotecan in colorectal carcinoma cells resistant to the single agents. Irinotecan 130-140 TNF receptor associated protein 1 Homo sapiens 48-53 19217207-6 2009 Conversely, the inhibition of TRAP1 activity by TRAP1 ATPase antagonist, shepherdin, increased the sensitivity to oxaliplatin and irinotecan in colorectal carcinoma cells resistant to the single agents. Irinotecan 130-140 dynein axonemal heavy chain 8 Homo sapiens 54-60 19201079-10 2009 The overexpression of ABCG2/BCRP was involved in the mechanism of resistance in SN-38-tolerant cells, and ABCC1/MRP1, ABCC3/MRP3, ABCC5/MRP5, and GGT1 may also have participated. Irinotecan 80-85 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 22-27 19201079-10 2009 The overexpression of ABCG2/BCRP was involved in the mechanism of resistance in SN-38-tolerant cells, and ABCC1/MRP1, ABCC3/MRP3, ABCC5/MRP5, and GGT1 may also have participated. Irinotecan 80-85 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 28-32 19528468-8 2009 TS protein level significantly decreased after exposure to SN-38. Irinotecan 59-64 thymidylate synthetase Homo sapiens 0-2 19528468-9 2009 CONCLUSION: The sequence dependency between SN-38 and 5-FU against colon cancer cells may be related to the dual action on cell cycle regulation by 5-FU and to the down-regulation of TS level by SN-38. Irinotecan 44-49 thymidylate synthetase Homo sapiens 183-185 19528468-9 2009 CONCLUSION: The sequence dependency between SN-38 and 5-FU against colon cancer cells may be related to the dual action on cell cycle regulation by 5-FU and to the down-regulation of TS level by SN-38. Irinotecan 195-200 thymidylate synthetase Homo sapiens 183-185 19581859-0 2009 Epidermal growth factor receptor expression discrepancies in metastatic colorectal cancer patients treated with cetuximab plus irinotecan-based chemotherapy refractory to irinotecan and oxaliplatin. Irinotecan 127-137 epidermal growth factor receptor Homo sapiens 0-32 18998135-2 2009 Irinotecan is used to treat a variety of solid tumours, through the inhibition of DNA topoisomerase I and is linked with severe mucositis and diarrhoea. Irinotecan 0-10 DNA topoisomerase I Rattus norvegicus 82-101 18998135-11 2009 CONCLUSIONS: Irinotecan causes an increase in mucin secretion and a net decrease in mucin-producing goblet cells, and the expression of Muc2 and Muc4 in the gastrointestinal tract is altered following treatment. Irinotecan 13-23 solute carrier family 13 member 2 Rattus norvegicus 46-51 18998135-11 2009 CONCLUSIONS: Irinotecan causes an increase in mucin secretion and a net decrease in mucin-producing goblet cells, and the expression of Muc2 and Muc4 in the gastrointestinal tract is altered following treatment. Irinotecan 13-23 solute carrier family 13 member 2 Rattus norvegicus 84-89 19581859-0 2009 Epidermal growth factor receptor expression discrepancies in metastatic colorectal cancer patients treated with cetuximab plus irinotecan-based chemotherapy refractory to irinotecan and oxaliplatin. Irinotecan 171-181 epidermal growth factor receptor Homo sapiens 0-32 19349540-7 2009 Almost 30% of the variability in SN-38 (the active metabolite of CPT-11) AUC is explained by ABCC1 1684T>C, ABCB1 IVS9 -44A>G, and UGT1A1*93 (P = .004). Irinotecan 65-71 ATP binding cassette subfamily C member 1 Homo sapiens 93-98 19398573-0 2009 PTEN expression and KRAS mutations on primary tumors and metastases in the prediction of benefit from cetuximab plus irinotecan for patients with metastatic colorectal cancer. Irinotecan 117-127 phosphatase and tensin homolog Homo sapiens 0-4 19398573-14 2009 CONCLUSION: PTEN loss in metastases may be predictive of resistance to cetuximab plus irinotecan. Irinotecan 86-96 phosphatase and tensin homolog Homo sapiens 12-16 19349540-7 2009 Almost 30% of the variability in SN-38 (the active metabolite of CPT-11) AUC is explained by ABCC1 1684T>C, ABCB1 IVS9 -44A>G, and UGT1A1*93 (P = .004). Irinotecan 65-71 ATP binding cassette subfamily B member 1 Homo sapiens 111-116 19349540-7 2009 Almost 30% of the variability in SN-38 (the active metabolite of CPT-11) AUC is explained by ABCC1 1684T>C, ABCB1 IVS9 -44A>G, and UGT1A1*93 (P = .004). Irinotecan 65-71 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 137-143 19349540-10 2009 CONCLUSION: On the basis of this exploratory analysis, common polymorphisms in genes encoding for ABC and SLC transporters may have a significant impact on the pharmacokinetics and pharmacodynamics of CPT-11. Irinotecan 201-207 ATP binding cassette subfamily B member 6 (Langereis blood group) Homo sapiens 98-101 19349540-10 2009 CONCLUSION: On the basis of this exploratory analysis, common polymorphisms in genes encoding for ABC and SLC transporters may have a significant impact on the pharmacokinetics and pharmacodynamics of CPT-11. Irinotecan 201-207 C-C motif chemokine ligand 21 Homo sapiens 106-109 19264971-6 2009 To determine the possible compensatory UGT isoforms, we first verified that UGT1A activities were significantly lower (p < 0.05) in Gunn rats by using UGT1A-specific probes 7-ethyl-10-hydroxycamptothecin (SN-38) and prunetin. Irinotecan 176-206 Ugt1a@ Rattus norvegicus 76-81 19264971-6 2009 To determine the possible compensatory UGT isoforms, we first verified that UGT1A activities were significantly lower (p < 0.05) in Gunn rats by using UGT1A-specific probes 7-ethyl-10-hydroxycamptothecin (SN-38) and prunetin. Irinotecan 208-213 Ugt1a@ Rattus norvegicus 76-81 19417717-0 2009 Effects of oral administration of S-1 on the pharmacokinetics of SN-38, irinotecan active metabolite, in patients with advanced colorectal cancer. Irinotecan 65-70 proteasome 26S subunit, non-ATPase 1 Homo sapiens 34-37 19349540-1 2009 PURPOSE: We aim to identify genetic variation, in addition to the UGT1A1*28 polymorphism, that can explain the variability in irinotecan (CPT-11) pharmacokinetics and neutropenia in cancer patients. Irinotecan 126-136 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 66-72 19349540-1 2009 PURPOSE: We aim to identify genetic variation, in addition to the UGT1A1*28 polymorphism, that can explain the variability in irinotecan (CPT-11) pharmacokinetics and neutropenia in cancer patients. Irinotecan 138-144 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 66-72 19349540-6 2009 More than 40% of the variation in CPT-11 area under the curve (AUC) is explained by ABCC2 -24C>T, SLCO1B1*5, HNF1A 79A>C, age, and CPT-11 dose (P < .0001). Irinotecan 34-40 ATP binding cassette subfamily C member 2 Homo sapiens 84-89 19349540-6 2009 More than 40% of the variation in CPT-11 area under the curve (AUC) is explained by ABCC2 -24C>T, SLCO1B1*5, HNF1A 79A>C, age, and CPT-11 dose (P < .0001). Irinotecan 34-40 solute carrier organic anion transporter family member 1B1 Homo sapiens 101-108 19349540-6 2009 More than 40% of the variation in CPT-11 area under the curve (AUC) is explained by ABCC2 -24C>T, SLCO1B1*5, HNF1A 79A>C, age, and CPT-11 dose (P < .0001). Irinotecan 34-40 HNF1 homeobox A Homo sapiens 112-117 19349540-7 2009 Almost 30% of the variability in SN-38 (the active metabolite of CPT-11) AUC is explained by ABCC1 1684T>C, ABCB1 IVS9 -44A>G, and UGT1A1*93 (P = .004). Irinotecan 33-38 ATP binding cassette subfamily C member 1 Homo sapiens 93-98 19349540-7 2009 Almost 30% of the variability in SN-38 (the active metabolite of CPT-11) AUC is explained by ABCC1 1684T>C, ABCB1 IVS9 -44A>G, and UGT1A1*93 (P = .004). Irinotecan 33-38 ATP binding cassette subfamily B member 1 Homo sapiens 111-116 19349540-7 2009 Almost 30% of the variability in SN-38 (the active metabolite of CPT-11) AUC is explained by ABCC1 1684T>C, ABCB1 IVS9 -44A>G, and UGT1A1*93 (P = .004). Irinotecan 33-38 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 137-143 19417717-0 2009 Effects of oral administration of S-1 on the pharmacokinetics of SN-38, irinotecan active metabolite, in patients with advanced colorectal cancer. Irinotecan 72-82 proteasome 26S subunit, non-ATPase 1 Homo sapiens 34-37 19417717-2 2009 The objective of this study was to describe the interaction between CPT-11 and S-1 in 4 patients with colorectal cancer. Irinotecan 68-74 proteasome 26S subunit, non-ATPase 1 Homo sapiens 79-82 19417717-3 2009 Coadministration of S-1 changed the pharmacokinetic behavior of CPT-11 and its metabolites. Irinotecan 64-70 proteasome 26S subunit, non-ATPase 1 Homo sapiens 20-23 19417717-4 2009 In particular, maximum plasma concentration (Cmax) and area under the plasma concentration curve (AUC) of 7-ethyl-10-hydroxycampothecin (SN-38) was markedly decreased by coadministration of S-1. Irinotecan 106-135 proteasome 26S subunit, non-ATPase 1 Homo sapiens 190-193 19417717-4 2009 In particular, maximum plasma concentration (Cmax) and area under the plasma concentration curve (AUC) of 7-ethyl-10-hydroxycampothecin (SN-38) was markedly decreased by coadministration of S-1. Irinotecan 137-142 proteasome 26S subunit, non-ATPase 1 Homo sapiens 190-193 19417717-5 2009 For SN-38, the median ratio of Cmax and AUC with S-1 to those without S-1 was median 0.34 (range 0.24-0.78) and 0.56 (range 0.23-0.68), respectively. Irinotecan 4-9 proteasome 26S subunit, non-ATPase 1 Homo sapiens 49-52 19417717-5 2009 For SN-38, the median ratio of Cmax and AUC with S-1 to those without S-1 was median 0.34 (range 0.24-0.78) and 0.56 (range 0.23-0.68), respectively. Irinotecan 4-9 proteasome 26S subunit, non-ATPase 1 Homo sapiens 70-73 19417717-7 2009 We conclude that the plasma concentration of SN-38 was decreased by oral administration of S-1 in patients with colorectal cancer. Irinotecan 45-50 proteasome 26S subunit, non-ATPase 1 Homo sapiens 91-94 19642458-5 2009 The anti-VEGFR bevacizumab is the standard first line treatment in metastatic colorectal cancer with irinotecan based chemotherapy. Irinotecan 101-111 kinase insert domain receptor Homo sapiens 9-14 19364970-1 2009 PURPOSE: UGT1A1*28 is considered the main pharmacogenetic predictor of the toxicity outcome of irinotecan-treated patients. Irinotecan 95-105 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 9-15 18998132-0 2009 Life-threatening toxicities in a patient with UGT1A1*6/*28 and SLCO1B1*15/*15 genotypes after irinotecan-based chemotherapy. Irinotecan 94-104 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 46-52 18998132-0 2009 Life-threatening toxicities in a patient with UGT1A1*6/*28 and SLCO1B1*15/*15 genotypes after irinotecan-based chemotherapy. Irinotecan 94-104 solute carrier organic anion transporter family member 1B1 Homo sapiens 63-70 18998132-6 2009 CONCLUSION: The severe toxicities in this patient are attributable to the extensive accumulation of SN-38, which may result from a synergistic or additive effect of low metabolic (UGT1A1*6/*28) and transport (SLCO1B1*15/*15) capabilities. Irinotecan 100-105 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 180-186 18998132-6 2009 CONCLUSION: The severe toxicities in this patient are attributable to the extensive accumulation of SN-38, which may result from a synergistic or additive effect of low metabolic (UGT1A1*6/*28) and transport (SLCO1B1*15/*15) capabilities. Irinotecan 100-105 solute carrier organic anion transporter family member 1B1 Homo sapiens 209-216 19461176-0 2009 [Evaluation of irinotecan hydrochloride (CPT-11) and trastuzumab combination therapy as salvage treatment in patients with HER2 overexpressing metastatic breast cancer]. Irinotecan 15-39 erb-b2 receptor tyrosine kinase 2 Homo sapiens 123-127 19461176-4 2009 PURPOSE: To evaluate retrospectively the efficacy of CPT-11 and trastuzumab combination therapy as salvage treatment in patients with human epidermal growth factor receptor 2 (HER2)overexpressing MBC. Irinotecan 53-59 erb-b2 receptor tyrosine kinase 2 Homo sapiens 140-174 19461176-4 2009 PURPOSE: To evaluate retrospectively the efficacy of CPT-11 and trastuzumab combination therapy as salvage treatment in patients with human epidermal growth factor receptor 2 (HER2)overexpressing MBC. Irinotecan 53-59 erb-b2 receptor tyrosine kinase 2 Homo sapiens 176-180 19450125-0 2009 UGT1A1*28 genotype predicts gastrointestinal toxicity in patients treated with intermediate-dose irinotecan. Irinotecan 97-107 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 19450125-1 2009 AIMS: Variants in UGT1A1 have previously been associated with toxicity from irinotecan chemotherapy. Irinotecan 76-86 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 18-24 19450125-2 2009 We conducted a pragmatic prospective cohort study to establish the relevance of UGT1A1 variants in the prediction of severe diarrhea and neutropenia in patients with colorectal cancer receiving irinotecan in a routine clinical setting. Irinotecan 194-204 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 80-86 19671328-8 2009 Cetuximab plus irinotecan as second-line therapy for patients with EGFR-expressing metastatic colorectal cancer who had previously failed to respond to irinotecan-added therapy could reach a tumor response rate of 16.4% - 23.0%, and median overall survival duration of 8.6 - 10.7 months. Irinotecan 15-25 epidermal growth factor receptor Homo sapiens 67-71 19225040-5 2009 Hydrolysis of imidapril and irinotecan hydrochloride (CPT-11) is catalyzed mainly by CES1 and CES2, respectively. Irinotecan 28-52 carboxylesterase 1 Homo sapiens 85-89 19225040-5 2009 Hydrolysis of imidapril and irinotecan hydrochloride (CPT-11) is catalyzed mainly by CES1 and CES2, respectively. Irinotecan 28-52 carboxylesterase 2 Homo sapiens 94-98 19225040-5 2009 Hydrolysis of imidapril and irinotecan hydrochloride (CPT-11) is catalyzed mainly by CES1 and CES2, respectively. Irinotecan 54-60 carboxylesterase 1 Homo sapiens 85-89 19225040-5 2009 Hydrolysis of imidapril and irinotecan hydrochloride (CPT-11) is catalyzed mainly by CES1 and CES2, respectively. Irinotecan 54-60 carboxylesterase 2 Homo sapiens 94-98 19390945-0 2009 UGT1A1*6 polymorphism is most predictive of severe neutropenia induced by irinotecan in Japanese cancer patients. Irinotecan 74-84 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 19262372-0 2009 Inhibition of P-glycoprotein-mediated docetaxel efflux sensitizes ovarian cancer cells to concomitant docetaxel and SN-38 exposure. Irinotecan 116-121 ATP binding cassette subfamily B member 1 Homo sapiens 14-28 19262372-8 2009 SN-38 increased P-gp expression in all cell lines. Irinotecan 0-5 ATP binding cassette subfamily B member 1 Homo sapiens 16-20 19390945-1 2009 BACKGROUND: Gene polymorphisms of the UDP-glucuronosyltransferase 1 family, polypeptide A1 (UGT1A1) contribute to individual variations in adverse events among patients administered irinotecan, and the distribution of the polymorphisms shows large interethnic differences. Irinotecan 182-192 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 38-90 19390945-1 2009 BACKGROUND: Gene polymorphisms of the UDP-glucuronosyltransferase 1 family, polypeptide A1 (UGT1A1) contribute to individual variations in adverse events among patients administered irinotecan, and the distribution of the polymorphisms shows large interethnic differences. Irinotecan 182-192 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 92-98 19287947-5 2009 Intraperitoneal injection of CPT-11, an anticancer topoisomerase I inhibitor that causes granulosa cell-specific apoptosis partly through Fas-Fas ligand (FasL) interactions in MCH mice, induced follicular apoptosis in both the wild-type and DAPK-mutant mice. Irinotecan 29-35 Fas ligand (TNF superfamily, member 6) Mus musculus 138-152 19390945-2 2009 Variation in the solute carrier organic anion-transporter family, member 1B1 (SLCO1B1) gene also has a significant effect on the disposition of irinotecan in Asian cancer patients. Irinotecan 144-154 solute carrier organic anion transporter family member 1B1 Homo sapiens 17-76 19287947-5 2009 Intraperitoneal injection of CPT-11, an anticancer topoisomerase I inhibitor that causes granulosa cell-specific apoptosis partly through Fas-Fas ligand (FasL) interactions in MCH mice, induced follicular apoptosis in both the wild-type and DAPK-mutant mice. Irinotecan 29-35 Fas ligand (TNF superfamily, member 6) Mus musculus 154-158 19287947-7 2009 The Fas and FasL expression levels in the CPT-11-injected mice did not differ significantly between the wild-type and DAPK-mutant mice. Irinotecan 42-48 Fas ligand (TNF superfamily, member 6) Mus musculus 12-16 19390945-2 2009 Variation in the solute carrier organic anion-transporter family, member 1B1 (SLCO1B1) gene also has a significant effect on the disposition of irinotecan in Asian cancer patients. Irinotecan 144-154 solute carrier organic anion transporter family member 1B1 Homo sapiens 78-85 19390945-12 2009 CONCLUSION: These findings suggest that the UGT1A1*6 polymorphism is a potential predictor of severe neutropenia caused by irinotecan in Japanese cancer patients. Irinotecan 123-133 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 44-50 19212674-3 2009 Although Akt activity seemed to be involved with the sensitivity of SCLC cells to chemotherapeutic agents (cisplatin, etoposide, SN38 and amrubicin), in Akt-activated N417 cells, only amrubicin exerted synergistic cell growth inhibition when combined with an Akt inhibitor, LY294002. Irinotecan 129-133 AKT serine/threonine kinase 1 Homo sapiens 9-12 19372567-5 2009 EJ human bladder carcinoma cells expressing membrane-tethered beta-glucuronidase (EJ/mbetaG cells) were used to selectively hydrolyze SN-38G to SN-38. Irinotecan 134-139 glucuronidase beta Homo sapiens 62-80 19372567-5 2009 EJ human bladder carcinoma cells expressing membrane-tethered beta-glucuronidase (EJ/mbetaG cells) were used to selectively hydrolyze SN-38G to SN-38. Irinotecan 144-149 glucuronidase beta Homo sapiens 62-80 19372567-11 2009 The high concentrations of SN-38G found in the serum of patients treated with CPT-11 suggest that clinical response to CPT-11 may be improved by elevating beta-glucuronidase activity in tumors. Irinotecan 27-32 glucuronidase beta Homo sapiens 155-173 19372567-11 2009 The high concentrations of SN-38G found in the serum of patients treated with CPT-11 suggest that clinical response to CPT-11 may be improved by elevating beta-glucuronidase activity in tumors. Irinotecan 78-84 glucuronidase beta Homo sapiens 155-173 19372567-11 2009 The high concentrations of SN-38G found in the serum of patients treated with CPT-11 suggest that clinical response to CPT-11 may be improved by elevating beta-glucuronidase activity in tumors. Irinotecan 119-125 glucuronidase beta Homo sapiens 155-173 19135530-0 2009 Effect of P-glycoprotein inhibitor, verapamil, on oral bioavailability and pharmacokinetics of irinotecan in rats. Irinotecan 95-105 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 10-24 19135530-1 2009 The objective of present investigation was to study the effect of verapamil on the pharmacokinetics of irinotecan in order to evaluate the role of P-glycoprotein (P-gp) in irinotecan disposition. Irinotecan 172-182 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 147-161 19135530-1 2009 The objective of present investigation was to study the effect of verapamil on the pharmacokinetics of irinotecan in order to evaluate the role of P-glycoprotein (P-gp) in irinotecan disposition. Irinotecan 172-182 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 163-167 19135530-5 2009 Bi-directional transport and inhibition studies in Caco-2 cells indicated irinotecan to be a P-gp substrate and the function of intestinal P-gp was significantly inhibited in presence of verapamil. Irinotecan 74-84 phosphoglycolate phosphatase Homo sapiens 93-97 19135530-5 2009 Bi-directional transport and inhibition studies in Caco-2 cells indicated irinotecan to be a P-gp substrate and the function of intestinal P-gp was significantly inhibited in presence of verapamil. Irinotecan 74-84 phosphoglycolate phosphatase Homo sapiens 139-143 19135530-13 2009 These results are quite stimulating for further development of a clinically useful oral formulation of irinotecan based on P-gp inhibition. Irinotecan 103-113 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 123-127 19212674-3 2009 Although Akt activity seemed to be involved with the sensitivity of SCLC cells to chemotherapeutic agents (cisplatin, etoposide, SN38 and amrubicin), in Akt-activated N417 cells, only amrubicin exerted synergistic cell growth inhibition when combined with an Akt inhibitor, LY294002. Irinotecan 129-133 AKT serine/threonine kinase 1 Homo sapiens 153-156 19212674-3 2009 Although Akt activity seemed to be involved with the sensitivity of SCLC cells to chemotherapeutic agents (cisplatin, etoposide, SN38 and amrubicin), in Akt-activated N417 cells, only amrubicin exerted synergistic cell growth inhibition when combined with an Akt inhibitor, LY294002. Irinotecan 129-133 AKT serine/threonine kinase 1 Homo sapiens 153-156 19190342-6 2009 Uptake of the organic cation [(3)H]MPP (4-methyl-pyridinium iodide) into these cells and also into hOCT3 stably transfected Chinese hamster ovary (CHO) cells was inhibited by irinotecan, vincristine, and melphalan. Irinotecan 175-185 solute carrier family 22 member 3 Homo sapiens 99-104 19190131-0 2009 Marked activity of irinotecan and rapamycin combination toward colon cancer cells in vivo and in vitro is mediated through cooperative modulation of the mammalian target of rapamycin/hypoxia-inducible factor-1alpha axis. Irinotecan 19-29 hypoxia inducible factor 1 subunit alpha Homo sapiens 183-214 19164213-4 2009 We investigated the association of FcgammaR polymorphisms and KRAS mutation with the outcome of irinotecan-refractory mCRC patients treated with cetuximab plus irinotecan. Irinotecan 96-106 KRAS proto-oncogene, GTPase Homo sapiens 62-66 19164213-11 2009 CONCLUSION: Combined FcgammaRIIa/FcgammaRIIIa polymorphisms are prognostic factors for disease progression in mCRC patients treated with cetuximab plus irinotecan. Irinotecan 152-162 Fc gamma receptor IIa Homo sapiens 21-32 19164213-11 2009 CONCLUSION: Combined FcgammaRIIa/FcgammaRIIIa polymorphisms are prognostic factors for disease progression in mCRC patients treated with cetuximab plus irinotecan. Irinotecan 152-162 Fc gamma receptor IIIa Homo sapiens 33-45 21475811-1 2009 This study aimed to clarify the mechanism by which apoptosis and Fas ligand (FasL) expression are induced in the ovarian granulosa cells of mice injected with irinotecan HCl (CPT-11). Irinotecan 159-173 Fas ligand (TNF superfamily, member 6) Mus musculus 65-75 21475811-1 2009 This study aimed to clarify the mechanism by which apoptosis and Fas ligand (FasL) expression are induced in the ovarian granulosa cells of mice injected with irinotecan HCl (CPT-11). Irinotecan 159-173 Fas ligand (TNF superfamily, member 6) Mus musculus 77-81 21475811-1 2009 This study aimed to clarify the mechanism by which apoptosis and Fas ligand (FasL) expression are induced in the ovarian granulosa cells of mice injected with irinotecan HCl (CPT-11). Irinotecan 175-181 Fas ligand (TNF superfamily, member 6) Mus musculus 65-75 21475811-1 2009 This study aimed to clarify the mechanism by which apoptosis and Fas ligand (FasL) expression are induced in the ovarian granulosa cells of mice injected with irinotecan HCl (CPT-11). Irinotecan 175-181 Fas ligand (TNF superfamily, member 6) Mus musculus 77-81 21475811-7 2009 In conclusion, apoptosis and FasL expression induced in the ovarian granulosa cells of mice injected with CPT-11 is not caused by direct stimulation with CPT-11 or SN38. Irinotecan 106-112 Fas ligand (TNF superfamily, member 6) Mus musculus 29-33 21475811-8 2009 Therefore, systemic CPT-11 administration appears to induce apoptosis and FasL expression in granulosa cells via currently unknown endogenous FasL-inducing factors or by active metabolites of CPT-11 other than SN38. Irinotecan 20-26 Fas ligand (TNF superfamily, member 6) Mus musculus 74-78 21475811-8 2009 Therefore, systemic CPT-11 administration appears to induce apoptosis and FasL expression in granulosa cells via currently unknown endogenous FasL-inducing factors or by active metabolites of CPT-11 other than SN38. Irinotecan 20-26 Fas ligand (TNF superfamily, member 6) Mus musculus 142-146 19190131-4 2009 A second class are topoisomerase I inhibitors, such as irinotecan, which are able to inhibit the accumulation of HIF-1alpha. Irinotecan 55-65 hypoxia inducible factor 1 subunit alpha Homo sapiens 113-123 19190131-11 2009 CONCLUSION: These results identify HIF-1alpha as a promising target and provide a rationale for clinical trials of low-dose irinotecan and rapamycin combination toward metastatic colon cancer. Irinotecan 124-134 hypoxia inducible factor 1 subunit alpha Homo sapiens 35-45 19190342-9 2009 The growth of CHO-hOCT3 was inhibited by 20% more with irinotecan and by 50% more with vincristine compared with nontransfected CHO cells. Irinotecan 55-65 solute carrier family 22 member 3 Homo sapiens 18-23 18685565-0 2009 Phase I/II pharmacokinetic and pharmacogenomic study of UGT1A1 polymorphism in elderly patients with advanced non-small cell lung cancer treated with irinotecan. Irinotecan 150-160 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 56-62 18929442-0 2009 Inhibition of Akt signaling by SN-38 induces apoptosis in cervical cancer. Irinotecan 31-36 AKT serine/threonine kinase 1 Homo sapiens 14-17 18929442-6 2009 Western Blot showed that SN-38 down-regulated protein expression of p-Akt and increased protein expression of p53 and p21, but it had no effects on protein expression of Bax, Bcl-2 and Akt. Irinotecan 25-30 AKT serine/threonine kinase 1 Homo sapiens 70-73 18929442-6 2009 Western Blot showed that SN-38 down-regulated protein expression of p-Akt and increased protein expression of p53 and p21, but it had no effects on protein expression of Bax, Bcl-2 and Akt. Irinotecan 25-30 tumor protein p53 Homo sapiens 110-113 18929442-6 2009 Western Blot showed that SN-38 down-regulated protein expression of p-Akt and increased protein expression of p53 and p21, but it had no effects on protein expression of Bax, Bcl-2 and Akt. Irinotecan 25-30 H3 histone pseudogene 16 Homo sapiens 118-121 18929442-7 2009 Transfection of the full-length Akt cDNA into HeLa and SiHa cells resulted in the reduction of apoptosis induced by SN-38, and Akt kinase activity regulated the p53 pathway, indicating that inhibition of the Akt pathway played an important role in exhibition of SN-38-mediated cytotoxic effect. Irinotecan 116-121 AKT serine/threonine kinase 1 Homo sapiens 32-35 18929442-7 2009 Transfection of the full-length Akt cDNA into HeLa and SiHa cells resulted in the reduction of apoptosis induced by SN-38, and Akt kinase activity regulated the p53 pathway, indicating that inhibition of the Akt pathway played an important role in exhibition of SN-38-mediated cytotoxic effect. Irinotecan 262-267 AKT serine/threonine kinase 1 Homo sapiens 32-35 18929442-7 2009 Transfection of the full-length Akt cDNA into HeLa and SiHa cells resulted in the reduction of apoptosis induced by SN-38, and Akt kinase activity regulated the p53 pathway, indicating that inhibition of the Akt pathway played an important role in exhibition of SN-38-mediated cytotoxic effect. Irinotecan 262-267 tumor protein p53 Homo sapiens 161-164 18929442-8 2009 Our data suggested that SN-38 could induce apoptosis through a p53 pathway and that activation of p53 in response to S-38 is governed by Akt. Irinotecan 24-29 tumor protein p53 Homo sapiens 63-66 18929442-8 2009 Our data suggested that SN-38 could induce apoptosis through a p53 pathway and that activation of p53 in response to S-38 is governed by Akt. Irinotecan 24-29 tumor protein p53 Homo sapiens 98-101 18685565-9 2009 An analysis of the influence of UGT1A1*28 and *6 polymorphisms provides useful information for the prediction of CPT-11-related hematological toxicity. Irinotecan 113-119 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 32-38 19208748-11 2009 Further, EFEMP1 expression decreased apoptosis and promoted cell cycle progression in response to serum starvation or exposure to gemcitabine, 5-fluorouracil, and irinotecan. Irinotecan 163-173 EGF containing fibulin extracellular matrix protein 1 Homo sapiens 9-15 18981166-0 2009 Close association of UGT1A9 IVS1+399C>T with UGT1A1*28, *6, or *60 haplotype and its apparent influence on 7-ethyl-10-hydroxycamptothecin (SN-38) glucuronidation in Japanese. Irinotecan 110-140 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 21-27 18981166-0 2009 Close association of UGT1A9 IVS1+399C>T with UGT1A1*28, *6, or *60 haplotype and its apparent influence on 7-ethyl-10-hydroxycamptothecin (SN-38) glucuronidation in Japanese. Irinotecan 142-147 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 21-27 18981166-0 2009 Close association of UGT1A9 IVS1+399C>T with UGT1A1*28, *6, or *60 haplotype and its apparent influence on 7-ethyl-10-hydroxycamptothecin (SN-38) glucuronidation in Japanese. Irinotecan 142-147 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 48-54 18981166-1 2009 The anticancer prodrug, irinotecan, is converted to its active form 7-ethyl-10-hydroxycamptothecin (SN-38) by carboxylesterases, and SN-38 is inactivated by UDP-glucuronosyltransferase (UGT)1A1-mediated glucuronidation. Irinotecan 24-34 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 186-193 18981166-1 2009 The anticancer prodrug, irinotecan, is converted to its active form 7-ethyl-10-hydroxycamptothecin (SN-38) by carboxylesterases, and SN-38 is inactivated by UDP-glucuronosyltransferase (UGT)1A1-mediated glucuronidation. Irinotecan 68-98 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 186-193 18981166-1 2009 The anticancer prodrug, irinotecan, is converted to its active form 7-ethyl-10-hydroxycamptothecin (SN-38) by carboxylesterases, and SN-38 is inactivated by UDP-glucuronosyltransferase (UGT)1A1-mediated glucuronidation. Irinotecan 100-105 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 186-193 18981166-1 2009 The anticancer prodrug, irinotecan, is converted to its active form 7-ethyl-10-hydroxycamptothecin (SN-38) by carboxylesterases, and SN-38 is inactivated by UDP-glucuronosyltransferase (UGT)1A1-mediated glucuronidation. Irinotecan 133-138 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 186-193 18981166-3 2009 In a recent study, it was reported that the UGT1A9 IVS1+399 (I399)C>T polymorphism is associated with increased SN-38 glucuronidation both in vitro and in vivo. Irinotecan 115-120 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 44-50 18981166-11 2009 Thus, at least in Japanese populations, influence of I399C>T on SN-38 glucuronidation is attributable to its close association with either UGT1A1*6, *28, or *60. Irinotecan 67-72 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 142-148 19234367-0 2009 Role of glucocorticoid receptor in the regulation of cellular sensitivity to irinotecan hydrochloride. Irinotecan 77-101 nuclear receptor subfamily 3, group C, member 1 Rattus norvegicus 8-31 19111841-1 2009 The breast cancer resistance protein, BCRP/ABCG2, is a half-molecule ATP-binding cassette transporter that facilitates the efflux of various anticancer agents from the cell, including 7-ethyl-10-hydroxycamptothecin, topotecan and mitoxantrone. Irinotecan 184-214 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 4-36 19111841-1 2009 The breast cancer resistance protein, BCRP/ABCG2, is a half-molecule ATP-binding cassette transporter that facilitates the efflux of various anticancer agents from the cell, including 7-ethyl-10-hydroxycamptothecin, topotecan and mitoxantrone. Irinotecan 184-214 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 38-42 19111841-1 2009 The breast cancer resistance protein, BCRP/ABCG2, is a half-molecule ATP-binding cassette transporter that facilitates the efflux of various anticancer agents from the cell, including 7-ethyl-10-hydroxycamptothecin, topotecan and mitoxantrone. Irinotecan 184-214 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 43-48 19118001-7 2009 In this drug-sensitive cellular background, MRP7 conferred high levels of resistance to docetaxel (46-fold), paclitaxel (116-fold), SN-38 (65-fold), daunorubicin (7.5-fold), etoposide (11-fold), and vincristine (56-fold). Irinotecan 132-137 ATP binding cassette subfamily C member 10 Homo sapiens 44-48 19147571-3 2009 We then report that chemotherapy induces Notch-1, as oxaliplatin, 5-fluorouracil (5-FU), or SN-38 (the active metabolite of irinotecan) induced Notch-1 intracellular domain (NICD) protein and activated Hes-1. Irinotecan 92-97 notch receptor 1 Homo sapiens 41-48 19147571-3 2009 We then report that chemotherapy induces Notch-1, as oxaliplatin, 5-fluorouracil (5-FU), or SN-38 (the active metabolite of irinotecan) induced Notch-1 intracellular domain (NICD) protein and activated Hes-1. Irinotecan 92-97 notch receptor 1 Homo sapiens 144-151 19147571-3 2009 We then report that chemotherapy induces Notch-1, as oxaliplatin, 5-fluorouracil (5-FU), or SN-38 (the active metabolite of irinotecan) induced Notch-1 intracellular domain (NICD) protein and activated Hes-1. Irinotecan 92-97 hes family bHLH transcription factor 1 Homo sapiens 202-207 19147571-3 2009 We then report that chemotherapy induces Notch-1, as oxaliplatin, 5-fluorouracil (5-FU), or SN-38 (the active metabolite of irinotecan) induced Notch-1 intracellular domain (NICD) protein and activated Hes-1. Irinotecan 124-134 notch receptor 1 Homo sapiens 41-48 19147571-3 2009 We then report that chemotherapy induces Notch-1, as oxaliplatin, 5-fluorouracil (5-FU), or SN-38 (the active metabolite of irinotecan) induced Notch-1 intracellular domain (NICD) protein and activated Hes-1. Irinotecan 124-134 notch receptor 1 Homo sapiens 144-151 19147571-3 2009 We then report that chemotherapy induces Notch-1, as oxaliplatin, 5-fluorouracil (5-FU), or SN-38 (the active metabolite of irinotecan) induced Notch-1 intracellular domain (NICD) protein and activated Hes-1. Irinotecan 124-134 hes family bHLH transcription factor 1 Homo sapiens 202-207 19147571-6 2009 Blocking the activation of Notch signaling with GSI34 sensitized cells to chemotherapy and was synergistic with oxaliplatin, 5-FU, and SN-38. Irinotecan 135-140 notch receptor 1 Homo sapiens 27-32 19414151-4 2009 UGT1A1*28 leads to reduced conjugation of SN-38, the active metabolite of irinotecan, resulting in an increased rate of adverse effects, especially neutropenia. Irinotecan 42-47 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 19414151-4 2009 UGT1A1*28 leads to reduced conjugation of SN-38, the active metabolite of irinotecan, resulting in an increased rate of adverse effects, especially neutropenia. Irinotecan 74-84 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 19208614-3 2009 Potent and specific inhibitors of PARP are available; these have been shown to increase the cytotoxicity of a range of anti-cancer agents including temozolomide, irinotecan and radiation. Irinotecan 162-172 poly(ADP-ribose) polymerase 1 Homo sapiens 34-38 19147776-1 2009 PURPOSE: CPX-1 is a novel, liposome-encapsulated formulation of irinotecan and floxuridine designed to prolong in vitro optimized synergistic molar ratios of both drugs postinfusion. Irinotecan 64-74 complexin 1 Homo sapiens 9-14 19151561-3 2009 To identify the optimal therapy candidates, EGFR (epidermal growth factor receptor)mutations are biomarkers available as predictive factors for EGFR tyrosine kinase inhibitors and UGT(uridine diphosphate glucuronosyltransferase)1A1 as a risk predictor of irinotecan in the present practice. Irinotecan 255-265 epidermal growth factor receptor Homo sapiens 44-48 19571434-10 2009 In the present study, Kampo was clarified to inhibit beta-estradiol and SN-38 glucuronidation mainly catalyzed by UGT1A1. Irinotecan 72-77 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 114-120 19151561-3 2009 To identify the optimal therapy candidates, EGFR (epidermal growth factor receptor)mutations are biomarkers available as predictive factors for EGFR tyrosine kinase inhibitors and UGT(uridine diphosphate glucuronosyltransferase)1A1 as a risk predictor of irinotecan in the present practice. Irinotecan 255-265 epidermal growth factor receptor Homo sapiens 50-82 19151561-3 2009 To identify the optimal therapy candidates, EGFR (epidermal growth factor receptor)mutations are biomarkers available as predictive factors for EGFR tyrosine kinase inhibitors and UGT(uridine diphosphate glucuronosyltransferase)1A1 as a risk predictor of irinotecan in the present practice. Irinotecan 255-265 epidermal growth factor receptor Homo sapiens 144-148 19151561-3 2009 To identify the optimal therapy candidates, EGFR (epidermal growth factor receptor)mutations are biomarkers available as predictive factors for EGFR tyrosine kinase inhibitors and UGT(uridine diphosphate glucuronosyltransferase)1A1 as a risk predictor of irinotecan in the present practice. Irinotecan 255-265 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 180-231 18221820-5 2009 The UGT1A1*6/*6, UGT1A9*1/*1 or *1/*22, and SLCO1B1 521TC or CC genotypes, and female-gender were predictive for higher AUC(SN-38) in multivariate analysis. Irinotecan 124-129 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 19125128-0 2009 Recommendations from the EGAPP Working Group: can UGT1A1 genotyping reduce morbidity and mortality in patients with metastatic colorectal cancer treated with irinotecan? Irinotecan 158-168 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 50-56 19125128-8 2009 CLINICAL VALIDITY: The EWG found adequate evidence of a significant association between UGT1A1 genotype and the incidence of severe neutropenia at standard doses of irinotecan. Irinotecan 165-175 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 88-94 19125128-16 2009 Further rigorous evaluation of UGT1A1 genotyping using current and promising irinotecan treatment protocols is warranted. Irinotecan 77-87 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 31-37 19125129-0 2009 Can UGT1A1 genotyping reduce morbidity and mortality in patients with metastatic colorectal cancer treated with irinotecan? Irinotecan 112-122 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 19125129-2 2009 This evidence-based review addresses the question of whether testing for UGT1A1 mutations in patients with metastatic colorectal cancer treated with irinotecan leads to improvement in outcomes (e.g., irinotecan toxicity, response to treatment, morbidity, and mortality), when compared with no testing. Irinotecan 149-159 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 73-79 19125129-2 2009 This evidence-based review addresses the question of whether testing for UGT1A1 mutations in patients with metastatic colorectal cancer treated with irinotecan leads to improvement in outcomes (e.g., irinotecan toxicity, response to treatment, morbidity, and mortality), when compared with no testing. Irinotecan 200-210 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 73-79 19125129-16 2009 Given the large number of colorectal cancer cases diagnosed each year, a randomized controlled trial of the effects of irinotecan dose modifications in patients with colorectal cancer based on their UGT1A1 genotype is feasible and could clarify the tradeoffs between possible reductions in severe neutropenia and improved tumor response and/or survival in patients with various UGT1A1 genotypes. Irinotecan 119-129 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 199-205 19125129-16 2009 Given the large number of colorectal cancer cases diagnosed each year, a randomized controlled trial of the effects of irinotecan dose modifications in patients with colorectal cancer based on their UGT1A1 genotype is feasible and could clarify the tradeoffs between possible reductions in severe neutropenia and improved tumor response and/or survival in patients with various UGT1A1 genotypes. Irinotecan 119-129 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 378-384 19404408-13 2009 In contrast, the MTD was determined to be dose level 4 (200 mg/m(2) for irinotecan and AUC 5 for carboplatin) by modified-Fibonacci simulation. Irinotecan 72-82 metallothionein 1E Homo sapiens 17-20 19534437-1 2009 BACKGROUND: Combination of cetuximab and irinotecan is an efficacious second- (and further-)-line treatment-alternative in patients with EGFR-positive metastatic colorectal cancer refractory to irinotecan. Irinotecan 41-51 epidermal growth factor receptor Homo sapiens 137-141 18221820-5 2009 The UGT1A1*6/*6, UGT1A9*1/*1 or *1/*22, and SLCO1B1 521TC or CC genotypes, and female-gender were predictive for higher AUC(SN-38) in multivariate analysis. Irinotecan 124-129 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 17-23 18221820-5 2009 The UGT1A1*6/*6, UGT1A9*1/*1 or *1/*22, and SLCO1B1 521TC or CC genotypes, and female-gender were predictive for higher AUC(SN-38) in multivariate analysis. Irinotecan 124-129 solute carrier organic anion transporter family member 1B1 Homo sapiens 44-51 19712005-3 2009 METHODS: Here, we assess the genome of 469 individuals from Sao Miguel Island (Azores, Portugal) in order to determine the frequencies of polymorphisms and haplotypes in UGT1A1, UGT1A6, and UGT1A7, the co-occurrence of reduced enzyme activity UGT1A variants related to irinotecan toxicity, and to calculate the extent of linkage disequilibrium (LD) in the genomic region encompassing these genes. Irinotecan 269-279 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 170-176 18820127-2 2009 Herein, we report that treatment with the 90-kDa heat shock protein (Hsp90) molecular chaperone inhibitor 17-allylamino-17-demethoxygeldanamycin (17AAG) selectively abrogates the G(2)/M checkpoint induced by 7-ethyl-10-hydroxycamptothecin (SN-38), an active metabolite of irinotecan, in p53-null compared with p53-intact HCT116 colon cancer cells. Irinotecan 208-238 heat shock protein 90 alpha family class A member 1 Homo sapiens 69-74 18820127-2 2009 Herein, we report that treatment with the 90-kDa heat shock protein (Hsp90) molecular chaperone inhibitor 17-allylamino-17-demethoxygeldanamycin (17AAG) selectively abrogates the G(2)/M checkpoint induced by 7-ethyl-10-hydroxycamptothecin (SN-38), an active metabolite of irinotecan, in p53-null compared with p53-intact HCT116 colon cancer cells. Irinotecan 208-238 N-methylpurine DNA glycosylase Homo sapiens 148-151 19712005-3 2009 METHODS: Here, we assess the genome of 469 individuals from Sao Miguel Island (Azores, Portugal) in order to determine the frequencies of polymorphisms and haplotypes in UGT1A1, UGT1A6, and UGT1A7, the co-occurrence of reduced enzyme activity UGT1A variants related to irinotecan toxicity, and to calculate the extent of linkage disequilibrium (LD) in the genomic region encompassing these genes. Irinotecan 269-279 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 170-175 18820127-2 2009 Herein, we report that treatment with the 90-kDa heat shock protein (Hsp90) molecular chaperone inhibitor 17-allylamino-17-demethoxygeldanamycin (17AAG) selectively abrogates the G(2)/M checkpoint induced by 7-ethyl-10-hydroxycamptothecin (SN-38), an active metabolite of irinotecan, in p53-null compared with p53-intact HCT116 colon cancer cells. Irinotecan 240-245 heat shock protein 90 alpha family class A member 1 Homo sapiens 69-74 18820127-2 2009 Herein, we report that treatment with the 90-kDa heat shock protein (Hsp90) molecular chaperone inhibitor 17-allylamino-17-demethoxygeldanamycin (17AAG) selectively abrogates the G(2)/M checkpoint induced by 7-ethyl-10-hydroxycamptothecin (SN-38), an active metabolite of irinotecan, in p53-null compared with p53-intact HCT116 colon cancer cells. Irinotecan 240-245 N-methylpurine DNA glycosylase Homo sapiens 148-151 19032367-1 2008 ATP-binding cassette transporter G2 (ABCG2) is the most recently described transporter of the multidrug-resistance pump and it promotes resistance to anticancer drugs such as doxorubicin, mitoxantrone, topotecan, and SN-38. Irinotecan 217-222 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 0-35 18820127-2 2009 Herein, we report that treatment with the 90-kDa heat shock protein (Hsp90) molecular chaperone inhibitor 17-allylamino-17-demethoxygeldanamycin (17AAG) selectively abrogates the G(2)/M checkpoint induced by 7-ethyl-10-hydroxycamptothecin (SN-38), an active metabolite of irinotecan, in p53-null compared with p53-intact HCT116 colon cancer cells. Irinotecan 272-282 heat shock protein 90 alpha family class A member 1 Homo sapiens 69-74 18820127-2 2009 Herein, we report that treatment with the 90-kDa heat shock protein (Hsp90) molecular chaperone inhibitor 17-allylamino-17-demethoxygeldanamycin (17AAG) selectively abrogates the G(2)/M checkpoint induced by 7-ethyl-10-hydroxycamptothecin (SN-38), an active metabolite of irinotecan, in p53-null compared with p53-intact HCT116 colon cancer cells. Irinotecan 272-282 N-methylpurine DNA glycosylase Homo sapiens 148-151 18820127-8 2009 Knockdown of Chk1 and Wee1 by short interfering RNA each resulted in abrogation of the G(2)/M checkpoint induced by SN-38. Irinotecan 116-121 checkpoint kinase 1 Homo sapiens 13-17 18820127-8 2009 Knockdown of Chk1 and Wee1 by short interfering RNA each resulted in abrogation of the G(2)/M checkpoint induced by SN-38. Irinotecan 116-121 WEE1 G2 checkpoint kinase Homo sapiens 22-26 18820127-9 2009 The combination of SN-38 and 17AAG was shown to be synergistic in p53-null but not in parental HCT116 cells by median effect/combination index analysis. Irinotecan 19-24 tumor protein p53 Homo sapiens 66-69 19738388-0 2009 The role of KRAS mutations in predicting the efficacy of cetuximab-plus-irinotecan therapy in irinotecan-refractory Korean metastatic colorectal cancer patients. Irinotecan 72-82 KRAS proto-oncogene, GTPase Homo sapiens 12-16 19738388-0 2009 The role of KRAS mutations in predicting the efficacy of cetuximab-plus-irinotecan therapy in irinotecan-refractory Korean metastatic colorectal cancer patients. Irinotecan 94-104 KRAS proto-oncogene, GTPase Homo sapiens 12-16 19738388-9 2009 CONCLUSIONS: This study indicates the clinical relevance of KRAS mutations in predicting the efficacy of cetuximab-plus-irinotecan-based chemotherapy in irinotecan-refractory Korean mCRC patients. Irinotecan 120-130 KRAS proto-oncogene, GTPase Homo sapiens 60-64 19738388-9 2009 CONCLUSIONS: This study indicates the clinical relevance of KRAS mutations in predicting the efficacy of cetuximab-plus-irinotecan-based chemotherapy in irinotecan-refractory Korean mCRC patients. Irinotecan 153-163 KRAS proto-oncogene, GTPase Homo sapiens 60-64 19299905-0 2009 Clinical significance of UDP-glucuronosyltransferase 1A1*6 for toxicities of combination chemotherapy with irinotecan and cisplatin in gynecologic cancers: a prospective multi-institutional study. Irinotecan 107-117 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 25-56 19299905-1 2009 BACKGROUND: To investigate the effects of UDP-glucuronosyltransferase 1A1 (UGT1A1) *28, *6 and *27 in patients with gynecologic cancer who received chemotherapy with irinotecan and cisplatin. Irinotecan 166-176 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 42-73 19299905-9 2009 CONCLUSION: In addition to UGT1A1*28, UGT1A1*6 might also be a key candidate to determine the dose of combination chemotherapy with irinotecan and cisplatin. Irinotecan 132-142 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 38-44 19366055-1 2009 BACKGROUND: Capecitabine in combination with oxaliplatin and irinotecan (COI regimen) is active and well tolerated in metastatic colorectal cancer. Irinotecan 61-71 mitochondrially encoded cytochrome c oxidase I Homo sapiens 73-76 19440006-0 2009 Treatment of gastric cancer cells with 5-fluorouracil/leucovorin and irinotecan induces distinct alterations in the mRNA expression of the apoptosis-related genes, including the novel gene BCL2L12. Irinotecan 69-79 BCL2 like 12 Homo sapiens 189-196 19440006-3 2009 The aim of this study was to investigate the modulations in the BAX, BCL2 and BCL2L12 mRNA levels in gastric cancer cells, after their treatment with the anticancer drugs 5-fluorouracil and irinotecan as well as the antioxidant substance leucovorin. Irinotecan 190-200 BCL2 associated X, apoptosis regulator Homo sapiens 64-67 19440006-3 2009 The aim of this study was to investigate the modulations in the BAX, BCL2 and BCL2L12 mRNA levels in gastric cancer cells, after their treatment with the anticancer drugs 5-fluorouracil and irinotecan as well as the antioxidant substance leucovorin. Irinotecan 190-200 BCL2 apoptosis regulator Homo sapiens 69-73 19440006-3 2009 The aim of this study was to investigate the modulations in the BAX, BCL2 and BCL2L12 mRNA levels in gastric cancer cells, after their treatment with the anticancer drugs 5-fluorouracil and irinotecan as well as the antioxidant substance leucovorin. Irinotecan 190-200 BCL2 like 12 Homo sapiens 78-85 19521145-6 2009 In combination therapy experiments, antibodies to PrP enhanced the effect of irinotecan, 5-FU, cisplatin and doxorubicin to varying degrees. Irinotecan 77-87 prion protein Homo sapiens 50-53 19088028-9 2008 CPT-11 treatment up-regulated Noxa expression, as did bortezomib, and enhanced Noxa/Mcl-1 complexes. Irinotecan 0-6 phorbol-12-myristate-13-acetate-induced protein 1 Homo sapiens 30-34 19088028-9 2008 CPT-11 treatment up-regulated Noxa expression, as did bortezomib, and enhanced Noxa/Mcl-1 complexes. Irinotecan 0-6 phorbol-12-myristate-13-acetate-induced protein 1 Homo sapiens 79-83 19088028-9 2008 CPT-11 treatment up-regulated Noxa expression, as did bortezomib, and enhanced Noxa/Mcl-1 complexes. Irinotecan 0-6 MCL1 apoptosis regulator, BCL2 family member Homo sapiens 84-89 19088028-10 2008 CPT-11 also disrupted the Mcl-1/Bak interaction. Irinotecan 0-6 MCL1 apoptosis regulator, BCL2 family member Homo sapiens 26-31 19088028-10 2008 CPT-11 also disrupted the Mcl-1/Bak interaction. Irinotecan 0-6 BCL2 antagonist/killer 1 Homo sapiens 32-35 19088028-11 2008 Knockdown of Noxa using short hairpin RNA lentiviral constructs was shown to significantly attenuate the cytotoxic effect of CPT-11 or bortezomib combined with ABT-737 and inhibited caspase-3 cleavage. Irinotecan 125-131 phorbol-12-myristate-13-acetate-induced protein 1 Homo sapiens 13-17 19088028-12 2008 CONCLUSIONS: Induction of Noxa by CPT-11 or bortezomib can sensitize colorectal cancer cells expressing Mcl-1 to ABT-737. Irinotecan 34-40 phorbol-12-myristate-13-acetate-induced protein 1 Homo sapiens 26-30 19088028-12 2008 CONCLUSIONS: Induction of Noxa by CPT-11 or bortezomib can sensitize colorectal cancer cells expressing Mcl-1 to ABT-737. Irinotecan 34-40 MCL1 apoptosis regulator, BCL2 family member Homo sapiens 104-109 18953066-0 2008 Genetic testing for UGT1A1*28 and *6 in Japanese patients who receive irinotecan chemotherapy. Irinotecan 70-80 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 20-26 18927500-0 2008 Faecal microflora and beta-glucuronidase expression are altered in an irinotecan-induced diarrhea model in rats. Irinotecan 70-80 glucuronidase, beta Rattus norvegicus 22-40 18927500-3 2008 Irinotecan causes cholinergic and delayed onset diarrhea in patients, in which beta-glucuronidase produced by gut bacteria is thought to be involved. Irinotecan 0-10 glucuronidase beta Homo sapiens 79-97 18927500-13 2008 CONCLUSIONS: Irinotecan-induced diarrhea may be caused by an increase in beta-glucuronidase producing bacteria. Irinotecan 13-23 glucuronidase, beta Rattus norvegicus 73-91 19032367-1 2008 ATP-binding cassette transporter G2 (ABCG2) is the most recently described transporter of the multidrug-resistance pump and it promotes resistance to anticancer drugs such as doxorubicin, mitoxantrone, topotecan, and SN-38. Irinotecan 217-222 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 37-42 19047118-1 2008 PURPOSE: Recently, an objective response rate of 12% was reported in a phase II study of cetuximab in patients with epidermal growth factor receptor (EGFR)-expressing metastatic colorectal cancer (mCRC) refractory to fluoropyrimidine-, oxaliplatin-, and irinotecan-based chemotherapy (IMC-0144). Irinotecan 254-264 epidermal growth factor receptor Homo sapiens 116-148 19047118-1 2008 PURPOSE: Recently, an objective response rate of 12% was reported in a phase II study of cetuximab in patients with epidermal growth factor receptor (EGFR)-expressing metastatic colorectal cancer (mCRC) refractory to fluoropyrimidine-, oxaliplatin-, and irinotecan-based chemotherapy (IMC-0144). Irinotecan 254-264 epidermal growth factor receptor Homo sapiens 150-154 19059062-3 2008 Irinotecan toxicity in patients who have colorectal cancer has been linked to reduced activity of uridine diphosphate-glucuronyltransferase 1A1 (UGT1A1). Irinotecan 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 98-143 19059062-3 2008 Irinotecan toxicity in patients who have colorectal cancer has been linked to reduced activity of uridine diphosphate-glucuronyltransferase 1A1 (UGT1A1). Irinotecan 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 145-151 18335219-1 2008 PURPOSE: A multicenter phase I trial to establish the recommended dose of CPT-11/docetaxel plus cisplatin in advanced esophagogastric cancer patients and to correlate the efficacy and toxicity with genetic polymorphisms in DNA repair genes (XPD and XRCC3) and the UGT1A1 gene. Irinotecan 74-80 ERCC excision repair 2, TFIIH core complex helicase subunit Homo sapiens 241-244 18981587-0 2008 Association of ATP-binding cassette, sub-family C, number 2 (ABCC2) genotype with pharmacokinetics of irinotecan in Japanese patients with metastatic colorectal cancer treated with irinotecan plus infusional 5-fluorouracil/leucovorin (FOLFIRI). Irinotecan 102-112 ATP binding cassette subfamily C member 2 Homo sapiens 15-59 18981587-0 2008 Association of ATP-binding cassette, sub-family C, number 2 (ABCC2) genotype with pharmacokinetics of irinotecan in Japanese patients with metastatic colorectal cancer treated with irinotecan plus infusional 5-fluorouracil/leucovorin (FOLFIRI). Irinotecan 102-112 ATP binding cassette subfamily C member 2 Homo sapiens 61-66 18981587-1 2008 ATP-binding cassette, sub-family C, number 2 (ABCC2) is involved in the biliary excretion of irinotecan and its metabolites, SN-38 and SN-38 glucuronide. Irinotecan 93-103 ATP binding cassette subfamily C member 2 Homo sapiens 0-44 18981587-1 2008 ATP-binding cassette, sub-family C, number 2 (ABCC2) is involved in the biliary excretion of irinotecan and its metabolites, SN-38 and SN-38 glucuronide. Irinotecan 93-103 ATP binding cassette subfamily C member 2 Homo sapiens 46-51 19028276-2 2008 In the present study, mRNA expression of PDGFRbeta and c-kit in 33 patients with locally advanced rectal cancer undergoing preoperative chemoradiotherapy with cetuximab/capecitabine/irinotecan in correlation with the tumor regression rate was investigated. Irinotecan 182-192 platelet derived growth factor receptor beta Homo sapiens 41-50 18941461-5 2008 hMLH1-deficient cell lines due to either epigenetic silencing or mutation showed very similar IC(50) and were four- to nine-fold more sensitive to CPT-11 than the MSS line. Irinotecan 147-153 mutL homolog 1 Homo sapiens 0-5 18941461-6 2008 Cell lines harbouring mutations in both MRE11 and RAD50 were most sensitive to CPT-11. Irinotecan 79-85 MRE11 homolog, double strand break repair nuclease Homo sapiens 40-45 18941461-6 2008 Cell lines harbouring mutations in both MRE11 and RAD50 were most sensitive to CPT-11. Irinotecan 79-85 RAD50 double strand break repair protein Homo sapiens 50-55 18981587-1 2008 ATP-binding cassette, sub-family C, number 2 (ABCC2) is involved in the biliary excretion of irinotecan and its metabolites, SN-38 and SN-38 glucuronide. Irinotecan 125-130 ATP binding cassette subfamily C member 2 Homo sapiens 0-44 18981587-1 2008 ATP-binding cassette, sub-family C, number 2 (ABCC2) is involved in the biliary excretion of irinotecan and its metabolites, SN-38 and SN-38 glucuronide. Irinotecan 125-130 ATP binding cassette subfamily C member 2 Homo sapiens 46-51 18981587-2 2008 Effects of the ABCC2 genotype on the pharmacokinetics (PK) of irinotecan and the metabolites were examined in Japanese patients with metastatic colorectal cancer receiving irinotecan plus infusional 5-fluorouracil/leucovorin (FOLFIRI). Irinotecan 62-72 ATP binding cassette subfamily C member 2 Homo sapiens 15-20 18981587-5 2008 Relationship between the ABCC2 genotypes or diplotypes and area under the time-concentration curve (AUC) of irinotecan and the metabolites normalized by irinotecan dose was analyzed. Irinotecan 108-118 ATP binding cassette subfamily C member 2 Homo sapiens 25-30 18981587-6 2008 The lower AUC of irinotecan was seen in patients with A/A or G/A genotypes at 1249 of the ABCC2 gene than others (p=0.011, Mann-Whitney U teat). Irinotecan 17-27 ATP binding cassette subfamily C member 2 Homo sapiens 90-95 18981587-12 2008 Thus, ABCC2 genotype is one of the predictors of the variability of irinotecan PK in Japanese patients with metastatic colorectal cancer receiving FOLFIRI. Irinotecan 68-78 ATP binding cassette subfamily C member 2 Homo sapiens 6-11 18278496-10 2008 RESULTS: The best structural model for irinotecan and its metabolites consisted of six-compartments: two compartments for irinotecan and SN-38, and one each for APC and SN-38G. Irinotecan 39-49 APC regulator of WNT signaling pathway Homo sapiens 161-164 18771527-0 2008 Overexpression of carboxylesterase-2 results in enhanced efficacy of topoisomerase I inhibitor, irinotecan (CPT-11), for multiple myeloma. Irinotecan 96-106 carboxylesterase 2 Homo sapiens 18-36 18771527-0 2008 Overexpression of carboxylesterase-2 results in enhanced efficacy of topoisomerase I inhibitor, irinotecan (CPT-11), for multiple myeloma. Irinotecan 108-114 carboxylesterase 2 Homo sapiens 18-36 18771527-5 2008 Interestingly, high expression of hCE-2 represented the nature of normal plasma cells, suggesting that hCE-2 could efficiently generate SN-38 within the plasma cells. Irinotecan 136-141 carboxylesterase 2 Homo sapiens 34-39 18771527-5 2008 Interestingly, high expression of hCE-2 represented the nature of normal plasma cells, suggesting that hCE-2 could efficiently generate SN-38 within the plasma cells. Irinotecan 136-141 carboxylesterase 2 Homo sapiens 103-108 18558463-6 2008 UGT1A1 *28 homozygosity is strongly associated with irinotecan-induced neutropenia and polymorphisms in the transporting peptides ABCB1 and OATP1B1 have also been associated with gastrointestinal toxicity and irinotecan pharmacokinetics, respectively. Irinotecan 52-62 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 18558463-6 2008 UGT1A1 *28 homozygosity is strongly associated with irinotecan-induced neutropenia and polymorphisms in the transporting peptides ABCB1 and OATP1B1 have also been associated with gastrointestinal toxicity and irinotecan pharmacokinetics, respectively. Irinotecan 209-219 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 18558463-6 2008 UGT1A1 *28 homozygosity is strongly associated with irinotecan-induced neutropenia and polymorphisms in the transporting peptides ABCB1 and OATP1B1 have also been associated with gastrointestinal toxicity and irinotecan pharmacokinetics, respectively. Irinotecan 209-219 ATP binding cassette subfamily B member 1 Homo sapiens 130-135 18558463-6 2008 UGT1A1 *28 homozygosity is strongly associated with irinotecan-induced neutropenia and polymorphisms in the transporting peptides ABCB1 and OATP1B1 have also been associated with gastrointestinal toxicity and irinotecan pharmacokinetics, respectively. Irinotecan 209-219 solute carrier organic anion transporter family member 1B1 Homo sapiens 140-147 18558463-7 2008 In the irinotecan product label, it is advised to reduce the irinotecan starting dose for UGT1A1 *28 homozygotes. Irinotecan 7-17 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 90-96 18558463-7 2008 In the irinotecan product label, it is advised to reduce the irinotecan starting dose for UGT1A1 *28 homozygotes. Irinotecan 61-71 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 90-96 18771527-6 2008 As expected, higher sensitivity to CPT-11 was observed in hCE-2-overexpressing U266 cells than mock U266 cells. Irinotecan 35-41 carboxylesterase 2 Homo sapiens 58-63 18335219-1 2008 PURPOSE: A multicenter phase I trial to establish the recommended dose of CPT-11/docetaxel plus cisplatin in advanced esophagogastric cancer patients and to correlate the efficacy and toxicity with genetic polymorphisms in DNA repair genes (XPD and XRCC3) and the UGT1A1 gene. Irinotecan 74-80 X-ray repair cross complementing 3 Homo sapiens 249-254 18335219-1 2008 PURPOSE: A multicenter phase I trial to establish the recommended dose of CPT-11/docetaxel plus cisplatin in advanced esophagogastric cancer patients and to correlate the efficacy and toxicity with genetic polymorphisms in DNA repair genes (XPD and XRCC3) and the UGT1A1 gene. Irinotecan 74-80 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 264-270 18801934-8 2008 Markers available for identifying drug-induced adverse reactions include thiopurine methyltransferase (TPMT) to predict toxicity from thiopurines in the treatment of acute lymphoblastic leukemia and uridine diphosphate glucuronyltransferase to predict toxicity from irinotecan in the treatment of colorectal cancer. Irinotecan 266-276 thiopurine S-methyltransferase Homo sapiens 103-107 19260480-0 2008 TACE of liver metastases from colorectal cancer adopting irinotecan-eluting beads: beneficial effect of palliative intra-arterial lidocaine and post-procedure supportive therapy on the control of side effects. Irinotecan 57-67 ADAM metallopeptidase domain 17 Homo sapiens 0-4 19004723-5 2008 Among the most conclusive examples one is that of the prediction of severe neutropenia induced by the irinotecan among patients homozygous for * 28 allele of UGT1A1 enzyme which conjugates SN38 active compound of irinotecan, the other one is the presence of a KRAS mutated allele in tumor cell to predict resistance to anti EGFR antibodies in the treatment of colorectal metastatic cancer. Irinotecan 102-112 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 158-164 18832463-3 2008 Homozygous carriers of UGT1A1*28 as well as those with additional UGT1A variants can suffer from severe irinotecan toxicity or jaundice during treatment with the protease inhibitor atazanavir. Irinotecan 104-114 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 23-29 18832463-3 2008 Homozygous carriers of UGT1A1*28 as well as those with additional UGT1A variants can suffer from severe irinotecan toxicity or jaundice during treatment with the protease inhibitor atazanavir. Irinotecan 104-114 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 23-28 18797455-0 2008 GSTP1 Ile105Val polymorphism correlates with progression-free survival in MCRC patients treated with or without irinotecan: a study of the Dutch Colorectal Cancer Group. Irinotecan 112-122 glutathione S-transferase pi 1 Homo sapiens 0-5 18797455-2 2008 As nuclear GSTP1 activity decreases irinotecan cytotoxicity, Val-allele carriers may benefit more from irinotecan chemotherapy. Irinotecan 36-46 glutathione S-transferase pi 1 Homo sapiens 11-16 18797455-2 2008 As nuclear GSTP1 activity decreases irinotecan cytotoxicity, Val-allele carriers may benefit more from irinotecan chemotherapy. Irinotecan 103-113 glutathione S-transferase pi 1 Homo sapiens 11-16 18797455-3 2008 Our aim was to investigate the association of GSTP1 genotype with treatment outcome of irinotecan. Irinotecan 87-97 glutathione S-transferase pi 1 Homo sapiens 46-51 18797455-10 2008 GSTP1 codon 105 polymorphism may be predictive for the response to irinotecan-based chemotherapy in patients with MCRC, with the Val-allele being associated with a better outcome. Irinotecan 67-77 glutathione S-transferase pi 1 Homo sapiens 0-5 18797458-2 2008 We included 49 patients treated for metastatic colorectal cancer with a combination of 5-fluorouracil and irinotecan; a polymorphism in the UGT1A1 gene (TA repeat in the TATA box) and one in the CES2 gene promoter (830C>G) were studied as potential markers for SN-38 glucuronidation and irinotecan activation, respectively; and the potential activity of CYP3A4 was estimated from cortisol biotransformation into 6beta-hydroxycortisol. Irinotecan 106-116 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 140-146 18797458-10 2008 UGT1A1 genotyping plus cortisol 6beta-hydroxylation determination could help to determine the optimal dose of irinotecan. Irinotecan 110-120 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 18927314-2 2008 We conducted a phase I study of irinotecan and the Hsp90 inhibitor 17AAG, which can also down-regulate Chk1, in patients with solid tumors. Irinotecan 32-42 checkpoint kinase 1 Homo sapiens 103-107 18720361-1 2008 Pharmacogenetic research indicates a relationship between a polymorphism in the gene encoding uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) and irinotecan inactivation, in that degradation of SN-38, the active metabolite of irinotecan, correlates inversely with the number of TA repeats in the TATA element of the UGT1A1 promoter region. Irinotecan 155-165 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 94-141 18720361-1 2008 Pharmacogenetic research indicates a relationship between a polymorphism in the gene encoding uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) and irinotecan inactivation, in that degradation of SN-38, the active metabolite of irinotecan, correlates inversely with the number of TA repeats in the TATA element of the UGT1A1 promoter region. Irinotecan 155-165 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 143-149 18720361-1 2008 Pharmacogenetic research indicates a relationship between a polymorphism in the gene encoding uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) and irinotecan inactivation, in that degradation of SN-38, the active metabolite of irinotecan, correlates inversely with the number of TA repeats in the TATA element of the UGT1A1 promoter region. Irinotecan 155-165 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 325-331 18720361-1 2008 Pharmacogenetic research indicates a relationship between a polymorphism in the gene encoding uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) and irinotecan inactivation, in that degradation of SN-38, the active metabolite of irinotecan, correlates inversely with the number of TA repeats in the TATA element of the UGT1A1 promoter region. Irinotecan 203-208 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 94-141 18720361-1 2008 Pharmacogenetic research indicates a relationship between a polymorphism in the gene encoding uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) and irinotecan inactivation, in that degradation of SN-38, the active metabolite of irinotecan, correlates inversely with the number of TA repeats in the TATA element of the UGT1A1 promoter region. Irinotecan 203-208 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 143-149 18720361-1 2008 Pharmacogenetic research indicates a relationship between a polymorphism in the gene encoding uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) and irinotecan inactivation, in that degradation of SN-38, the active metabolite of irinotecan, correlates inversely with the number of TA repeats in the TATA element of the UGT1A1 promoter region. Irinotecan 203-208 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 325-331 18720361-1 2008 Pharmacogenetic research indicates a relationship between a polymorphism in the gene encoding uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) and irinotecan inactivation, in that degradation of SN-38, the active metabolite of irinotecan, correlates inversely with the number of TA repeats in the TATA element of the UGT1A1 promoter region. Irinotecan 235-245 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 94-141 18720361-1 2008 Pharmacogenetic research indicates a relationship between a polymorphism in the gene encoding uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) and irinotecan inactivation, in that degradation of SN-38, the active metabolite of irinotecan, correlates inversely with the number of TA repeats in the TATA element of the UGT1A1 promoter region. Irinotecan 235-245 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 143-149 18720361-1 2008 Pharmacogenetic research indicates a relationship between a polymorphism in the gene encoding uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) and irinotecan inactivation, in that degradation of SN-38, the active metabolite of irinotecan, correlates inversely with the number of TA repeats in the TATA element of the UGT1A1 promoter region. Irinotecan 235-245 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 325-331 18720361-2 2008 Individuals who are homozygous for the UGT1A1*28 allele (7 repeats) may exhibit reduced degradation of SN-38 and increased probability of severe toxicities. Irinotecan 103-108 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 39-45 18720361-5 2008 Irinotecan labeling recommends testing for the UGT1A1*28 allele and reducing irinotecan dosing in patients who are positive to reduce the likelihood of dose-limiting neutropenia only, but not diarrhea. Irinotecan 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 47-53 18703021-7 2008 A number of Pgp substrates (quinidine, amprenavir, irinotecan, topotecan, atorvastatin and erythromycin) induced net digoxin secretion, as did the non-Pgp substrate artemisinin. Irinotecan 51-61 ATP binding cassette subfamily B member 1 Homo sapiens 12-15 19004723-5 2008 Among the most conclusive examples one is that of the prediction of severe neutropenia induced by the irinotecan among patients homozygous for * 28 allele of UGT1A1 enzyme which conjugates SN38 active compound of irinotecan, the other one is the presence of a KRAS mutated allele in tumor cell to predict resistance to anti EGFR antibodies in the treatment of colorectal metastatic cancer. Irinotecan 102-112 KRAS proto-oncogene, GTPase Homo sapiens 260-264 19004723-5 2008 Among the most conclusive examples one is that of the prediction of severe neutropenia induced by the irinotecan among patients homozygous for * 28 allele of UGT1A1 enzyme which conjugates SN38 active compound of irinotecan, the other one is the presence of a KRAS mutated allele in tumor cell to predict resistance to anti EGFR antibodies in the treatment of colorectal metastatic cancer. Irinotecan 102-112 epidermal growth factor receptor Homo sapiens 324-328 19004723-5 2008 Among the most conclusive examples one is that of the prediction of severe neutropenia induced by the irinotecan among patients homozygous for * 28 allele of UGT1A1 enzyme which conjugates SN38 active compound of irinotecan, the other one is the presence of a KRAS mutated allele in tumor cell to predict resistance to anti EGFR antibodies in the treatment of colorectal metastatic cancer. Irinotecan 213-223 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 158-164 19004723-5 2008 Among the most conclusive examples one is that of the prediction of severe neutropenia induced by the irinotecan among patients homozygous for * 28 allele of UGT1A1 enzyme which conjugates SN38 active compound of irinotecan, the other one is the presence of a KRAS mutated allele in tumor cell to predict resistance to anti EGFR antibodies in the treatment of colorectal metastatic cancer. Irinotecan 213-223 KRAS proto-oncogene, GTPase Homo sapiens 260-264 19004723-5 2008 Among the most conclusive examples one is that of the prediction of severe neutropenia induced by the irinotecan among patients homozygous for * 28 allele of UGT1A1 enzyme which conjugates SN38 active compound of irinotecan, the other one is the presence of a KRAS mutated allele in tumor cell to predict resistance to anti EGFR antibodies in the treatment of colorectal metastatic cancer. Irinotecan 213-223 epidermal growth factor receptor Homo sapiens 324-328 18794444-3 2008 Recent studies show that imatinib reverses ABCG2-mediated drug resistance to topotecan hydrochloride and SN-38. Irinotecan 105-110 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 43-48 18418374-7 2008 Polymorphisms in genes UGT1A1, UGT1A7, and UGT1A9 had strong associations with a component corresponding to the irinotecan-to-SN-38 pathway and SN-38 recirculation and to a component relating to SN-38-to-SN-38G conversion and elimination of SN-38G. Irinotecan 112-122 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 23-29 18418374-7 2008 Polymorphisms in genes UGT1A1, UGT1A7, and UGT1A9 had strong associations with a component corresponding to the irinotecan-to-SN-38 pathway and SN-38 recirculation and to a component relating to SN-38-to-SN-38G conversion and elimination of SN-38G. Irinotecan 144-149 UDP glucuronosyltransferase family 1 member A7 Homo sapiens 31-37 18765539-0 2008 Thymidine selectively enhances growth suppressive effects of camptothecin/irinotecan in MSI+ cells and tumors containing a mutation of MRE11. Irinotecan 74-84 phenylalkylamine Ca2+ antagonist (emopamil) binding protein Mus musculus 88-91 18765539-10 2008 Increased sensitivity to this combination is also evident in vivo as thymidine enhances irinotecan-induced growth suppression of MMR-deficient tumors carrying the MRE11 mutation in mouse xenografts. Irinotecan 88-98 MRE11A homolog A, double strand break repair nuclease Mus musculus 163-168 18765539-11 2008 CONCLUSION: Irinotecan-thymidine combinations may be particularly effective when targeted to MSI+ tumors containing this readily detectable MRE11 mutation. Irinotecan 12-22 phenylalkylamine Ca2+ antagonist (emopamil) binding protein Mus musculus 93-96 18765539-11 2008 CONCLUSION: Irinotecan-thymidine combinations may be particularly effective when targeted to MSI+ tumors containing this readily detectable MRE11 mutation. Irinotecan 12-22 MRE11A homolog A, double strand break repair nuclease Mus musculus 140-145 18418374-7 2008 Polymorphisms in genes UGT1A1, UGT1A7, and UGT1A9 had strong associations with a component corresponding to the irinotecan-to-SN-38 pathway and SN-38 recirculation and to a component relating to SN-38-to-SN-38G conversion and elimination of SN-38G. Irinotecan 144-149 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 43-49 18418374-7 2008 Polymorphisms in genes UGT1A1, UGT1A7, and UGT1A9 had strong associations with a component corresponding to the irinotecan-to-SN-38 pathway and SN-38 recirculation and to a component relating to SN-38-to-SN-38G conversion and elimination of SN-38G. Irinotecan 144-149 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 23-29 18418374-7 2008 Polymorphisms in genes UGT1A1, UGT1A7, and UGT1A9 had strong associations with a component corresponding to the irinotecan-to-SN-38 pathway and SN-38 recirculation and to a component relating to SN-38-to-SN-38G conversion and elimination of SN-38G. Irinotecan 112-122 UDP glucuronosyltransferase family 1 member A7 Homo sapiens 31-37 18418374-7 2008 Polymorphisms in genes UGT1A1, UGT1A7, and UGT1A9 had strong associations with a component corresponding to the irinotecan-to-SN-38 pathway and SN-38 recirculation and to a component relating to SN-38-to-SN-38G conversion and elimination of SN-38G. Irinotecan 144-149 UDP glucuronosyltransferase family 1 member A7 Homo sapiens 31-37 18418374-7 2008 Polymorphisms in genes UGT1A1, UGT1A7, and UGT1A9 had strong associations with a component corresponding to the irinotecan-to-SN-38 pathway and SN-38 recirculation and to a component relating to SN-38-to-SN-38G conversion and elimination of SN-38G. Irinotecan 144-149 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 43-49 18418374-7 2008 Polymorphisms in genes UGT1A1, UGT1A7, and UGT1A9 had strong associations with a component corresponding to the irinotecan-to-SN-38 pathway and SN-38 recirculation and to a component relating to SN-38-to-SN-38G conversion and elimination of SN-38G. Irinotecan 112-122 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 43-49 18418374-8 2008 The component characterizing irinotecan"s compartments was associated with HNF1alpha and ABCC2 polymorphisms. Irinotecan 29-39 HNF1 homeobox A Homo sapiens 75-84 18418374-7 2008 Polymorphisms in genes UGT1A1, UGT1A7, and UGT1A9 had strong associations with a component corresponding to the irinotecan-to-SN-38 pathway and SN-38 recirculation and to a component relating to SN-38-to-SN-38G conversion and elimination of SN-38G. Irinotecan 126-131 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 23-29 18418374-8 2008 The component characterizing irinotecan"s compartments was associated with HNF1alpha and ABCC2 polymorphisms. Irinotecan 29-39 ATP binding cassette subfamily C member 2 Homo sapiens 89-94 18418374-7 2008 Polymorphisms in genes UGT1A1, UGT1A7, and UGT1A9 had strong associations with a component corresponding to the irinotecan-to-SN-38 pathway and SN-38 recirculation and to a component relating to SN-38-to-SN-38G conversion and elimination of SN-38G. Irinotecan 126-131 UDP glucuronosyltransferase family 1 member A7 Homo sapiens 31-37 18418374-7 2008 Polymorphisms in genes UGT1A1, UGT1A7, and UGT1A9 had strong associations with a component corresponding to the irinotecan-to-SN-38 pathway and SN-38 recirculation and to a component relating to SN-38-to-SN-38G conversion and elimination of SN-38G. Irinotecan 126-131 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 43-49 18418374-7 2008 Polymorphisms in genes UGT1A1, UGT1A7, and UGT1A9 had strong associations with a component corresponding to the irinotecan-to-SN-38 pathway and SN-38 recirculation and to a component relating to SN-38-to-SN-38G conversion and elimination of SN-38G. Irinotecan 144-149 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 23-29 18418374-7 2008 Polymorphisms in genes UGT1A1, UGT1A7, and UGT1A9 had strong associations with a component corresponding to the irinotecan-to-SN-38 pathway and SN-38 recirculation and to a component relating to SN-38-to-SN-38G conversion and elimination of SN-38G. Irinotecan 144-149 UDP glucuronosyltransferase family 1 member A7 Homo sapiens 31-37 18418374-7 2008 Polymorphisms in genes UGT1A1, UGT1A7, and UGT1A9 had strong associations with a component corresponding to the irinotecan-to-SN-38 pathway and SN-38 recirculation and to a component relating to SN-38-to-SN-38G conversion and elimination of SN-38G. Irinotecan 144-149 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 43-49 18418374-7 2008 Polymorphisms in genes UGT1A1, UGT1A7, and UGT1A9 had strong associations with a component corresponding to the irinotecan-to-SN-38 pathway and SN-38 recirculation and to a component relating to SN-38-to-SN-38G conversion and elimination of SN-38G. Irinotecan 144-149 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 23-29 18418374-7 2008 Polymorphisms in genes UGT1A1, UGT1A7, and UGT1A9 had strong associations with a component corresponding to the irinotecan-to-SN-38 pathway and SN-38 recirculation and to a component relating to SN-38-to-SN-38G conversion and elimination of SN-38G. Irinotecan 144-149 UDP glucuronosyltransferase family 1 member A7 Homo sapiens 31-37 18418374-7 2008 Polymorphisms in genes UGT1A1, UGT1A7, and UGT1A9 had strong associations with a component corresponding to the irinotecan-to-SN-38 pathway and SN-38 recirculation and to a component relating to SN-38-to-SN-38G conversion and elimination of SN-38G. Irinotecan 144-149 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 43-49 18418374-7 2008 Polymorphisms in genes UGT1A1, UGT1A7, and UGT1A9 had strong associations with a component corresponding to the irinotecan-to-SN-38 pathway and SN-38 recirculation and to a component relating to SN-38-to-SN-38G conversion and elimination of SN-38G. Irinotecan 144-149 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 23-29 18619980-1 2008 P-glycoprotein (P-gp) is found to play a very significant role in intestinal and biliary transport of irinotecan and its active metabolite, SN-38. Irinotecan 102-112 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 0-14 18509065-5 2008 In particular, SN-38 arrested cells in S phase, enhanced the accumulation of gemcitabine metabolites, and diminished checkpoint kinase 1, thereby sensitizing cells in the SN-38 --> gemcitabine sequence. Irinotecan 15-20 checkpoint kinase 1 Homo sapiens 117-136 18695900-10 2008 The results from the present subcutaneously implanted tumor model suggested that a higher efficacy may be expected when bevacizumab is combined with the cytotoxic agent CPT-11, compared to bevacizumab alone, against tumors with a variety of VEGF production levels in clinical situations. Irinotecan 169-175 vascular endothelial growth factor A Homo sapiens 241-245 18619980-1 2008 P-glycoprotein (P-gp) is found to play a very significant role in intestinal and biliary transport of irinotecan and its active metabolite, SN-38. Irinotecan 102-112 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 16-20 18619980-1 2008 P-glycoprotein (P-gp) is found to play a very significant role in intestinal and biliary transport of irinotecan and its active metabolite, SN-38. Irinotecan 140-145 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 0-14 18619980-1 2008 P-glycoprotein (P-gp) is found to play a very significant role in intestinal and biliary transport of irinotecan and its active metabolite, SN-38. Irinotecan 140-145 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 16-20 18619980-2 2008 This makes P-gp inhibition a logical strategy for improving irinotecan"s oral efficacy and reducing its toxicity. Irinotecan 60-70 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 11-15 18619980-10 2008 Our studies not only propose a safe approach for bioavailability enhancement and reducing toxicity of irinotecan by P-gp inhibition but in another way also reiterate the significance of elucidating herb-drug interactions for future insights. Irinotecan 102-112 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 116-120 17992531-7 2008 RESULTS: Area under the concentration-time curve ratios of APC/irinotecan, an in vivo parameter for CYP3A4 activity, were significantly higher in females than in males. Irinotecan 63-73 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 100-106 18816295-2 2008 UGT1A9 is a UGT that catalyses the conjugation of endogenous oestrogenic and thyroid hormones, acetaminophen, SN-38 (an active metabolite of irinotecan) and phenols. Irinotecan 110-115 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 0-6 18816295-2 2008 UGT1A9 is a UGT that catalyses the conjugation of endogenous oestrogenic and thyroid hormones, acetaminophen, SN-38 (an active metabolite of irinotecan) and phenols. Irinotecan 141-151 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 0-6 18579105-9 2008 The blood to bile distribution ratio (AUC(bile)/AUC(blood)) was significantly reduced after group coadministration of EGCG, it can be seen that the bile efflux transport system of CPT-11 and SN-38 may be markedly reduced by the treatment of EGCG which plays the role of P-gp inhibitor. Irinotecan 180-186 ATP binding cassette subfamily B member 1 Homo sapiens 270-274 18579105-9 2008 The blood to bile distribution ratio (AUC(bile)/AUC(blood)) was significantly reduced after group coadministration of EGCG, it can be seen that the bile efflux transport system of CPT-11 and SN-38 may be markedly reduced by the treatment of EGCG which plays the role of P-gp inhibitor. Irinotecan 191-196 ATP binding cassette subfamily B member 1 Homo sapiens 270-274 17992531-0 2008 Impact of CYP3A4 haplotypes on irinotecan pharmacokinetics in Japanese cancer patients. Irinotecan 31-41 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 10-16 17992531-1 2008 BACKGROUND AND PURPOSE: Cytochrome P450 3A4 (CYP3A4) converts an anticancer prodrug, irinotecan, to inactive metabolites such as APC. Irinotecan 85-95 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 24-43 17992531-1 2008 BACKGROUND AND PURPOSE: Cytochrome P450 3A4 (CYP3A4) converts an anticancer prodrug, irinotecan, to inactive metabolites such as APC. Irinotecan 85-95 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 45-51 17992531-12 2008 CONCLUSION: This study suggested that CYP3A4*16B was associated with decreased metabolism of irinotecan to APC. Irinotecan 93-103 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 38-44 17992531-2 2008 However, the contribution of CYP3A4 genetic polymorphisms to irinotecan pharmacokinetics (PK) and pharmacodynamics (PD) is not fully elucidated. Irinotecan 61-71 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 29-35 18690847-3 2008 The aim of the study was to investigate the induction by celecoxib of some multidrug resistance proteins, MRP1, MRP2, MRP4 and MRP5, involved in the transport of irinotecan and 5-FU. Irinotecan 162-172 ATP binding cassette subfamily C member 1 Homo sapiens 106-110 17992531-4 2008 In this study, the effects of CYP3A4 haplotypes including *16B on irinotecan PK/PD were investigated in irinotecan-administered patients. Irinotecan 66-76 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 30-36 17992531-5 2008 METHODS: The CYP3A4 genotypes for 177 Japanese cancer patients who received irinotecan were defined in terms of 4 major haplotypes, i.e., *1A (wild type), *1G (IVS10 + 12G > A), *16B [554C > G (Thr185Ser) and IVS10 + 12G > A], and *18B [878T > C (Leu293Pro) and IVS10 + 12G > A]. Irinotecan 76-86 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 13-19 17992531-6 2008 Associations of CYP3A4 genotypes with irinotecan PK and severe toxicities (grade 3 diarrhea and grade 3 or 4 neutropenia) were investigated. Irinotecan 38-48 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 16-22 18690847-3 2008 The aim of the study was to investigate the induction by celecoxib of some multidrug resistance proteins, MRP1, MRP2, MRP4 and MRP5, involved in the transport of irinotecan and 5-FU. Irinotecan 162-172 ATP binding cassette subfamily C member 2 Homo sapiens 112-116 18690847-3 2008 The aim of the study was to investigate the induction by celecoxib of some multidrug resistance proteins, MRP1, MRP2, MRP4 and MRP5, involved in the transport of irinotecan and 5-FU. Irinotecan 162-172 ATP binding cassette subfamily C member 4 Homo sapiens 118-122 18690847-3 2008 The aim of the study was to investigate the induction by celecoxib of some multidrug resistance proteins, MRP1, MRP2, MRP4 and MRP5, involved in the transport of irinotecan and 5-FU. Irinotecan 162-172 ATP binding cassette subfamily C member 5 Homo sapiens 127-131 18633245-0 2008 [Role of UGT1A1*28 and UGT1A1*6 for irinotecan-induced adverse drug reaction]. Irinotecan 36-46 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 9-15 18594531-1 2008 The aim of the study was to investigate the associations between UGT1A1(*)28 genotype and (1) response rates, (2) febrile neutropenia and (3) dose intensity in patients with metastatic colorectal cancer treated with irinotecan. Irinotecan 216-226 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 65-71 18594531-2 2008 UGT1A1(*)28 genotype was determined in 218 patients receiving irinotecan (either first-line therapy with capecitabine or second-line as monotherapy) for metastatic colorectal cancer. Irinotecan 62-72 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 18632807-0 2008 Colorectal tumor cells treated with 5-FU, oxaliplatin, irinotecan, and cetuximab exhibit changes in 18F-FDG incorporation corresponding to hexokinase activity and glucose transport. Irinotecan 55-65 hexokinase 1 Homo sapiens 139-149 18708359-7 2008 Resistance of TuBEC to chemotherapeutic agents such as CPT-11, etoposide, and temozolomide can be overcome by knockdown of GRP78 using small interfering RNA or chemical inhibition of its catalytic site. Irinotecan 55-61 heat shock protein family A (Hsp70) member 5 Homo sapiens 123-128 19343150-8 2008 In a randomized trial comparing 5-FU vs. irinotecan plus cisplatin vs. S-1, conducted by the Japan Clinical Oncology Group, S-1 was associated with a favorable safety profile, response rate, and time-to-treatment failure with longer survival compared with 5-FU. Irinotecan 41-51 proteasome 26S subunit, non-ATPase 1 Homo sapiens 124-127 18633245-0 2008 [Role of UGT1A1*28 and UGT1A1*6 for irinotecan-induced adverse drug reaction]. Irinotecan 36-46 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 23-29 18633245-3 2008 Irinotecan is metabolized by carboxylesterase to form an active metabolite, 7-ethyl-10-hydroxycamptothecin(SN-38), which in turn is subsequently conjugated by UGT-glucuronosyltransferase 1A1(UGT1A1)to yield an inactive form, SN-38 glucuronide(SN-38 G). Irinotecan 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 191-197 18633245-3 2008 Irinotecan is metabolized by carboxylesterase to form an active metabolite, 7-ethyl-10-hydroxycamptothecin(SN-38), which in turn is subsequently conjugated by UGT-glucuronosyltransferase 1A1(UGT1A1)to yield an inactive form, SN-38 glucuronide(SN-38 G). Irinotecan 76-106 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 191-197 18633245-3 2008 Irinotecan is metabolized by carboxylesterase to form an active metabolite, 7-ethyl-10-hydroxycamptothecin(SN-38), which in turn is subsequently conjugated by UGT-glucuronosyltransferase 1A1(UGT1A1)to yield an inactive form, SN-38 glucuronide(SN-38 G). Irinotecan 107-112 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 191-197 18633245-4 2008 The UGT1A1 gene polymorphisms contribute to the individual variation in adverse events among patients administered irinotecan. Irinotecan 115-125 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 18633245-7 2008 Recently, it has been demonstrated that genetic variants of UGT1A1*6 in addition to UGT1A1*28 are associated with the occurrence of adverse events in irinotecan chemotherapy in Asians. Irinotecan 150-160 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 60-66 18633245-7 2008 Recently, it has been demonstrated that genetic variants of UGT1A1*6 in addition to UGT1A1*28 are associated with the occurrence of adverse events in irinotecan chemotherapy in Asians. Irinotecan 150-160 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 84-90 18633245-8 2008 This review summarizes recent studies to outline the role of UGT1A1*28 and UGT1A1*6 for irinotecan-induced adverse drug reaction in Japanese cancer patients. Irinotecan 88-98 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 61-67 18633245-8 2008 This review summarizes recent studies to outline the role of UGT1A1*28 and UGT1A1*6 for irinotecan-induced adverse drug reaction in Japanese cancer patients. Irinotecan 88-98 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 75-81 18609706-11 2008 Furthermore, cells with reduced Bcl-x(L) or Mcl-1 expression was more sensitive towards oxaliplatin- and irinotecan-induced apoptosis, and in the case of Bcl-x(L) also towards 5-FU-induced apoptosis. Irinotecan 105-115 BCL2 like 1 Homo sapiens 32-40 18320294-1 2008 PURPOSE: The inter/intramolecular interactions between drugs (floxuridine, irinotecan) and excipients (copper gluconate, triethanolamine) in the dual-drug liposomal formulation CPX-1 were elucidated in order to identify the physicochemical properties that allow coordinated release of irinotecan and floxuridine and maintenance of the two agents at a fixed, synergistic 1:1 molar ratio. Irinotecan 75-85 complexin 1 Homo sapiens 177-182 18320294-5 2008 Approximately 45% of the total irinotecan was detectable in the copper-containing CPX-1 formulation by NMR, which decreased to 19% without copper present in the liposomal interior. Irinotecan 31-41 complexin 1 Homo sapiens 82-87 18609706-11 2008 Furthermore, cells with reduced Bcl-x(L) or Mcl-1 expression was more sensitive towards oxaliplatin- and irinotecan-induced apoptosis, and in the case of Bcl-x(L) also towards 5-FU-induced apoptosis. Irinotecan 105-115 MCL1 apoptosis regulator, BCL2 family member Homo sapiens 44-49 18384433-0 2008 Vascular endothelial growth factor expression is closely related to irinotecan-mediated inhibition of tumor growth and angiogenesis in neuroblastoma xenografts. Irinotecan 68-78 vascular endothelial growth factor A Mus musculus 0-34 17703303-3 2008 The aim of this study was to determine the expression of nuclear factor-kappaB (NF-kappaB), tumor necrosis factor (TNF) and interleukins-1beta (IL-1beta) and -6 (IL-6) in the AT following the administration of the chemotherapeutic agent irinotecan. Irinotecan 237-247 interleukin 6 Rattus norvegicus 162-166 18384433-8 2008 Our data demonstrate that diminished VEGF gene and protein expression is closely correlated with tumor growth inhibition and inhibition of angiogenesis by CPT-11 in NB xenografts. Irinotecan 155-161 vascular endothelial growth factor A Mus musculus 37-41 18633223-1 2008 Irinotecan hydrochloride is an inhibitor of DNA topoisomerase I enzyme by its main active metabolite SN-38. Irinotecan 0-24 DNA topoisomerase I Rattus norvegicus 44-63 18445521-7 2008 In conclusion, ST1968 exhibited an outstanding antitumour activity superior to that of irinotecan against SCC. Irinotecan 87-97 serpin family B member 3 Homo sapiens 106-109 18633223-1 2008 Irinotecan hydrochloride is an inhibitor of DNA topoisomerase I enzyme by its main active metabolite SN-38. Irinotecan 101-106 DNA topoisomerase I Rattus norvegicus 44-63 18633225-1 2008 We encountered a 64-year-old UGT1A1*28 heterotype patient who received CPT-11 and CDDP chemotherapy for Stage III A small-cell lung-cancer and developed grade 4 neutropenia as assessed by CTCAE v3.0 in Pharmaceutical Management. Irinotecan 71-77 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 29-35 18503403-5 2008 By performing uridine diphosphate glucuronosyltransferase (UGT) 1A1 genetic testing, some studies have been able to predict which patients receiving irinotecan will experience the toxicity. Irinotecan 149-159 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 14-67 18497997-8 2008 Furthermore, quantitative real-time PCR demonstrated that mRNA changes of selected novel genes (CENPB, H2AFX, MCM5, ZADH1 and NGB) in irinotecan-resistant clones vs. parental clone were in agreement with array-CGH results. Irinotecan 134-144 centromere protein B Homo sapiens 96-101 18497997-8 2008 Furthermore, quantitative real-time PCR demonstrated that mRNA changes of selected novel genes (CENPB, H2AFX, MCM5, ZADH1 and NGB) in irinotecan-resistant clones vs. parental clone were in agreement with array-CGH results. Irinotecan 134-144 H2A.X variant histone Homo sapiens 103-108 18497997-8 2008 Furthermore, quantitative real-time PCR demonstrated that mRNA changes of selected novel genes (CENPB, H2AFX, MCM5, ZADH1 and NGB) in irinotecan-resistant clones vs. parental clone were in agreement with array-CGH results. Irinotecan 134-144 minichromosome maintenance complex component 5 Homo sapiens 110-114 18497997-8 2008 Furthermore, quantitative real-time PCR demonstrated that mRNA changes of selected novel genes (CENPB, H2AFX, MCM5, ZADH1 and NGB) in irinotecan-resistant clones vs. parental clone were in agreement with array-CGH results. Irinotecan 134-144 prostaglandin reductase 2 Homo sapiens 116-121 18497997-8 2008 Furthermore, quantitative real-time PCR demonstrated that mRNA changes of selected novel genes (CENPB, H2AFX, MCM5, ZADH1 and NGB) in irinotecan-resistant clones vs. parental clone were in agreement with array-CGH results. Irinotecan 134-144 neuroglobin Homo sapiens 126-129 17700594-0 2008 Influence of UGT1A9 intronic I399C>T polymorphism on SN-38 glucuronidation in Asian cancer patients. Irinotecan 56-61 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 13-19 17700594-1 2008 Genetic polymorphisms in hepatically expressed UGT1A1 and UGT1A9 contribute to the interindividual variability i-n irinotecan disposition and toxicity. Irinotecan 115-125 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 47-53 17700594-1 2008 Genetic polymorphisms in hepatically expressed UGT1A1 and UGT1A9 contribute to the interindividual variability i-n irinotecan disposition and toxicity. Irinotecan 115-125 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 58-64 17700594-5 2008 UGT1A1-1A9 diplotype analysis showed that patients harbouring the H1/H2 (TG6GT(10)T/GG6GT(9)C) diplotype had 2.4-fold lower systemic exposure to SN-38 glucuronide (SN-38G) compared with patients harbouring the H1/H5 (TG6GT(10)T/GG6GT(10)C) diplotype (P=0.025). Irinotecan 145-150 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 17700594-7 2008 and suggests that the UGT1A9 I399C>T variant may be an important glucuronidating allele affecting the pharmacokinetics of SN-38 and SN-38G in Asian cancer patients receiving irinotecan chemotherapy. Irinotecan 125-130 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 22-28 17700594-7 2008 and suggests that the UGT1A9 I399C>T variant may be an important glucuronidating allele affecting the pharmacokinetics of SN-38 and SN-38G in Asian cancer patients receiving irinotecan chemotherapy. Irinotecan 135-140 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 22-28 17700594-7 2008 and suggests that the UGT1A9 I399C>T variant may be an important glucuronidating allele affecting the pharmacokinetics of SN-38 and SN-38G in Asian cancer patients receiving irinotecan chemotherapy. Irinotecan 177-187 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 22-28 17943230-1 2008 Clinically relevant resistance to the currently approved camptothecins, irinotecan and topotecan, is poorly understood but may involve increased expression of ATP-dependent drug transporters such as ABCG2 (breast cancer resistant protein, BCRP). Irinotecan 72-82 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 199-204 17943230-1 2008 Clinically relevant resistance to the currently approved camptothecins, irinotecan and topotecan, is poorly understood but may involve increased expression of ATP-dependent drug transporters such as ABCG2 (breast cancer resistant protein, BCRP). Irinotecan 72-82 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 239-243 18172616-1 2008 Human UDP-glucuronosyltransferase (UGT)1A1 is a critical enzyme responsible for detoxification and metabolism of endogenous and exogenous lipophilic compounds, such as potentially neurotoxic bilirubin and the anticancer drug irinotecan SN-38, via conjugation with glucuronic acid. Irinotecan 225-235 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 6-42 18172616-1 2008 Human UDP-glucuronosyltransferase (UGT)1A1 is a critical enzyme responsible for detoxification and metabolism of endogenous and exogenous lipophilic compounds, such as potentially neurotoxic bilirubin and the anticancer drug irinotecan SN-38, via conjugation with glucuronic acid. Irinotecan 236-241 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 6-42 18652401-4 2008 Polymorphisms of UGT 1A1 are considered as potential indicators of a risk of toxicity treatment by irinotecan. Irinotecan 99-109 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 17-24 17624531-7 2008 RESULTS: CPT-11 caused significant diarrhea, histopathological alterations (inflammatory cell infiltration, loss of crypt architecture and villus shortening) and increased intestinal tissue MPO activity, TNF-alpha, IL-1beta and KC level and TNF-alpha immuno-staining. Irinotecan 9-15 tumor necrosis factor Mus musculus 241-250 18443598-6 2008 SN-38 preferentially inhibited endothelial cell proliferation alone and interacted synergistically with semaxinib; it induced apoptosis and increased the expression and secretion of TSP-1. Irinotecan 0-5 thrombospondin 1 Homo sapiens 182-187 18443598-7 2008 Metronomic CPT-11 alone and combined with semaxinib significantly inhibits tumour growth in the absence of toxicity, which was accompanied by decreases in microvessel density and increases in TSP-1 gene expression in tumour tissues. Irinotecan 11-17 thrombospondin 1 Homo sapiens 192-197 18443598-8 2008 In vitro results show the antiangiogenic properties of low-concentration SN-38, suggesting a key role of TSP-1 in this effect. Irinotecan 73-78 thrombospondin 1 Homo sapiens 105-110 18472966-6 2008 Therapy with PKI166 alone or with irinotecan produced apoptosis of these endothelial cells and necrosis of the EGFR-negative tumors. Irinotecan 34-44 epidermal growth factor receptor Homo sapiens 111-115 18248513-6 2008 Both of naringenin and elacridar separately enhanced the sensitivity for CPT-11 and SN-38 in KYN-2 cells abundantly expressing BCRP, CYP3A4/5 and UGT1A1, but not in KYN-1 cells expressing lower levels. Irinotecan 73-79 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 127-131 18248513-6 2008 Both of naringenin and elacridar separately enhanced the sensitivity for CPT-11 and SN-38 in KYN-2 cells abundantly expressing BCRP, CYP3A4/5 and UGT1A1, but not in KYN-1 cells expressing lower levels. Irinotecan 84-89 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 127-131 18248513-6 2008 Both of naringenin and elacridar separately enhanced the sensitivity for CPT-11 and SN-38 in KYN-2 cells abundantly expressing BCRP, CYP3A4/5 and UGT1A1, but not in KYN-1 cells expressing lower levels. Irinotecan 84-89 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 133-139 18248513-6 2008 Both of naringenin and elacridar separately enhanced the sensitivity for CPT-11 and SN-38 in KYN-2 cells abundantly expressing BCRP, CYP3A4/5 and UGT1A1, but not in KYN-1 cells expressing lower levels. Irinotecan 84-89 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 146-152 18248513-8 2008 On the other hand, naringenin and elacridar significantly increased the chemosensitivity for CPT-11 and SN-38 in the KYN-1-derived cells artificially overexpressing BCRP. Irinotecan 93-99 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 165-169 18248513-8 2008 On the other hand, naringenin and elacridar significantly increased the chemosensitivity for CPT-11 and SN-38 in the KYN-1-derived cells artificially overexpressing BCRP. Irinotecan 104-109 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 165-169 18248513-10 2008 Those results suggest that BCRP is one of the chemosensitivity determinants of CPT-11 in HCC cells and its inhibition might be critical for cells expressing abundant BCRP. Irinotecan 79-85 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 27-31 18248513-10 2008 Those results suggest that BCRP is one of the chemosensitivity determinants of CPT-11 in HCC cells and its inhibition might be critical for cells expressing abundant BCRP. Irinotecan 79-85 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 166-170 17624531-1 2008 INTRODUCTION: Irinotecan (CPT-11) is an inhibitor of DNA topoisomerase I and is clinically effective against several cancers. Irinotecan 14-24 topoisomerase (DNA) I Mus musculus 53-72 17624531-1 2008 INTRODUCTION: Irinotecan (CPT-11) is an inhibitor of DNA topoisomerase I and is clinically effective against several cancers. Irinotecan 26-32 topoisomerase (DNA) I Mus musculus 53-72 17624531-7 2008 RESULTS: CPT-11 caused significant diarrhea, histopathological alterations (inflammatory cell infiltration, loss of crypt architecture and villus shortening) and increased intestinal tissue MPO activity, TNF-alpha, IL-1beta and KC level and TNF-alpha immuno-staining. Irinotecan 9-15 myeloperoxidase Mus musculus 190-193 17624531-7 2008 RESULTS: CPT-11 caused significant diarrhea, histopathological alterations (inflammatory cell infiltration, loss of crypt architecture and villus shortening) and increased intestinal tissue MPO activity, TNF-alpha, IL-1beta and KC level and TNF-alpha immuno-staining. Irinotecan 9-15 tumor necrosis factor Mus musculus 204-213 17624531-7 2008 RESULTS: CPT-11 caused significant diarrhea, histopathological alterations (inflammatory cell infiltration, loss of crypt architecture and villus shortening) and increased intestinal tissue MPO activity, TNF-alpha, IL-1beta and KC level and TNF-alpha immuno-staining. Irinotecan 9-15 interleukin 1 beta Mus musculus 215-223 18541046-12 2008 We also estimates the potential effects of genetic variations: the AUC and Cmax of SN-38 may increase to 208% and 165% respectively with the genetic variation UGT1A1*28/*28 which reduces the expression of UGT1A1 down to 30%. Irinotecan 83-88 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 159-165 18541046-12 2008 We also estimates the potential effects of genetic variations: the AUC and Cmax of SN-38 may increase to 208% and 165% respectively with the genetic variation UGT1A1*28/*28 which reduces the expression of UGT1A1 down to 30%. Irinotecan 83-88 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 205-211 18300238-0 2008 UGT1A1*28 polymorphism predicts irinotecan-induced severe toxicities without affecting treatment outcome and survival in patients with metastatic colorectal carcinoma. Irinotecan 32-42 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 18300238-1 2008 BACKGROUND: It is known that the uridine-diphosphoglucuronosyl transferase 1A1 (UGT1A1)*28 polymorphism reduces UGT1A1 enzyme activity, which may lead to severe toxicities in patients who receive irinotecan. Irinotecan 196-206 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-78 18300238-1 2008 BACKGROUND: It is known that the uridine-diphosphoglucuronosyl transferase 1A1 (UGT1A1)*28 polymorphism reduces UGT1A1 enzyme activity, which may lead to severe toxicities in patients who receive irinotecan. Irinotecan 196-206 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 80-86 18300238-1 2008 BACKGROUND: It is known that the uridine-diphosphoglucuronosyl transferase 1A1 (UGT1A1)*28 polymorphism reduces UGT1A1 enzyme activity, which may lead to severe toxicities in patients who receive irinotecan. Irinotecan 196-206 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 112-118 18300238-11 2008 CONCLUSIONS: The current data suggested that the UGT1A1*28 polymorphism may be a key determinant for predicting irinotecan-induced severe toxicities without affecting treatment outcome for patients with metastatic CRC. Irinotecan 112-122 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 49-55 18425325-0 2008 A novel molecular mechanism for anticancer drug-induced ovarian failure: Irinotecan HCl, an anticancer topoisomerase I inhibitor, induces specific FasL expression in granulosa cells of large ovarian follicles to enhance follicular apoptosis. Irinotecan 73-87 Fas ligand (TNF superfamily, member 6) Mus musculus 147-151 18425325-3 2008 Clinically therapeutic doses of CPT-11 were injected intraperitoneally into 8-week-old female MCH mice, and their ovaries were examined by the TUNEL assay to detect dead cells. Irinotecan 32-38 modifier of chinchilla Mus musculus 94-97 18425325-6 2008 Intraperitoneal CPT-11 injections induced specific TUNEL-positive cells and cell death with cleaved caspase 3 expression among large ovarian follicular granulosa cells. Irinotecan 16-22 caspase 3 Mus musculus 100-109 18425325-11 2008 Although no FasL expression was detected in normal ovarian tissues, CPT-11 injections significantly induced specific FasL expression in granulosa cells. Irinotecan 68-74 Fas ligand (TNF superfamily, member 6) Mus musculus 117-121 18425325-13 2008 In conclusion, CPT-11 dose-dependently induced specific FasL expression in granulosa cells of developing ovarian follicles. Irinotecan 15-21 Fas ligand (TNF superfamily, member 6) Mus musculus 56-60 18437170-4 2008 In addition, the Food and Drug Administration of the United States now requires a "black box" warning on the packaging of irinotecan for evaluation of germline polymorphism in UGT1A1, the gene mutated in Gilbert"s syndrome, for potential reduction of drug dosage in patients with the UGT1A1*28 polymorphism. Irinotecan 122-132 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 176-182 18437170-4 2008 In addition, the Food and Drug Administration of the United States now requires a "black box" warning on the packaging of irinotecan for evaluation of germline polymorphism in UGT1A1, the gene mutated in Gilbert"s syndrome, for potential reduction of drug dosage in patients with the UGT1A1*28 polymorphism. Irinotecan 122-132 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 284-290 18466101-0 2008 Cost-effectiveness of UGT1A1 genotyping in second-line, high-dose, once every 3 weeks irinotecan monotherapy treatment of colorectal cancer. Irinotecan 86-96 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 22-28 18466101-1 2008 AIM: The aim of the present study was to evaluate the cost-effectiveness of UGT1A1 genotyping in second-line, high-dose, once every 3 weeks irinotecan monotherapy treatment of colorectal cancer. Irinotecan 140-150 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 76-82 18466101-5 2008 RESULTS & CONCLUSION: Genotyping in combination with a subsequent reduction of initial irinotecan dose for UGT1A1 7/7 genotype patients was cost-saving for the population of African and Caucasian origin. Irinotecan 91-101 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 111-117 18466101-8 2008 Finally, the application of a 3-weekly high-dose treatment regimen with a 20% reduced dosage compared with the low-dose weekly irinotecan regimen in patients with UGT1A1 7/7 genotype was less expensive and is more convenient for the patient. Irinotecan 127-137 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 163-169 17549477-0 2008 Involvement of UDP-glucuronosyltransferase activity in irinotecan-induced delayed-onset diarrhea in rats. Irinotecan 55-65 UDP glycosyltransferase 2 family, polypeptide B Rattus norvegicus 15-42 17549477-1 2008 We assessed the involvement of UDP-glucuronosyltransferase (UGT) activity in episodes of irinotecan hydrochloride (CPT-11)-induced delayed-onset diarrhea using a mutant rat strain with an inherited deficiency of UGT1A (Gunn rats). Irinotecan 89-113 UDP glycosyltransferase 2 family, polypeptide B Rattus norvegicus 31-58 17549477-1 2008 We assessed the involvement of UDP-glucuronosyltransferase (UGT) activity in episodes of irinotecan hydrochloride (CPT-11)-induced delayed-onset diarrhea using a mutant rat strain with an inherited deficiency of UGT1A (Gunn rats). Irinotecan 89-113 UDP glycosyltransferase 2 family, polypeptide B Rattus norvegicus 60-63 17549477-1 2008 We assessed the involvement of UDP-glucuronosyltransferase (UGT) activity in episodes of irinotecan hydrochloride (CPT-11)-induced delayed-onset diarrhea using a mutant rat strain with an inherited deficiency of UGT1A (Gunn rats). Irinotecan 115-121 UDP glycosyltransferase 2 family, polypeptide B Rattus norvegicus 31-58 17549477-1 2008 We assessed the involvement of UDP-glucuronosyltransferase (UGT) activity in episodes of irinotecan hydrochloride (CPT-11)-induced delayed-onset diarrhea using a mutant rat strain with an inherited deficiency of UGT1A (Gunn rats). Irinotecan 115-121 UDP glycosyltransferase 2 family, polypeptide B Rattus norvegicus 60-63 17549477-10 2008 These results clarified that the deficiency of UGT activity contributed greatly to the induction of the CPT-11-induced delayed-onset diarrhea and epithelial impairment in the intestine. Irinotecan 104-110 UDP glycosyltransferase 2 family, polypeptide B Rattus norvegicus 47-50 17549477-11 2008 In the clinic, great care is needed when using chemotherapy with CPT-11 in patients with poor UGT activity. Irinotecan 65-71 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 94-97 18381438-3 2008 With the combination therapy of SN-38 (the active metabolite of CPT-11) followed by flavopiridol, the induction of apoptosis was 5-fold greater in the p53+/+ cells compared with the p53-/- cells. Irinotecan 64-70 tumor protein p53 Homo sapiens 151-154 18381438-3 2008 With the combination therapy of SN-38 (the active metabolite of CPT-11) followed by flavopiridol, the induction of apoptosis was 5-fold greater in the p53+/+ cells compared with the p53-/- cells. Irinotecan 64-70 tumor protein p53 Homo sapiens 182-185 18381438-3 2008 With the combination therapy of SN-38 (the active metabolite of CPT-11) followed by flavopiridol, the induction of apoptosis was 5-fold greater in the p53+/+ cells compared with the p53-/- cells. Irinotecan 32-37 tumor protein p53 Homo sapiens 151-154 17624531-10 2008 CONCLUSION: These results suggest an important role of TNF-alpha, IL-1beta and KC in the pathogenesis of intestinal mucositis induced by CPT-11. Irinotecan 137-143 tumor necrosis factor Mus musculus 55-64 18381438-3 2008 With the combination therapy of SN-38 (the active metabolite of CPT-11) followed by flavopiridol, the induction of apoptosis was 5-fold greater in the p53+/+ cells compared with the p53-/- cells. Irinotecan 32-37 tumor protein p53 Homo sapiens 182-185 18381438-7 2008 Depletion of Rad51 by small interfering RNA (siRNA) sensitized both p53+/+ and p53-/- cells to SN-38-induced apoptosis with increase of gamma H2AX, a marker of DNA damage. Irinotecan 95-100 RAD51 recombinase Homo sapiens 13-18 18381438-7 2008 Depletion of Rad51 by small interfering RNA (siRNA) sensitized both p53+/+ and p53-/- cells to SN-38-induced apoptosis with increase of gamma H2AX, a marker of DNA damage. Irinotecan 95-100 tumor protein p53 Homo sapiens 79-82 17624531-10 2008 CONCLUSION: These results suggest an important role of TNF-alpha, IL-1beta and KC in the pathogenesis of intestinal mucositis induced by CPT-11. Irinotecan 137-143 interleukin 1 beta Mus musculus 66-74 18082937-0 2008 Importance of UDP-glucuronosyltransferase 1A1*6 for irinotecan toxicities in Japanese cancer patients. Irinotecan 52-62 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 14-45 18467239-0 2008 UGT1A1*28 and other UGT1A polymorphisms as determinants of irinotecan toxicity. Irinotecan 59-69 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 18467239-0 2008 UGT1A1*28 and other UGT1A polymorphisms as determinants of irinotecan toxicity. Irinotecan 59-69 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 0-5 18413798-9 2008 The antitumor effect of RGD-L-TRAIL was further enhanced by combination with CPT-11 in both TRAIL-sensitive COLO-205 and TRAIL-resistive HT-29 tumor xenograft models. Irinotecan 77-83 tumor necrosis factor (ligand) superfamily, member 10 Mus musculus 30-35 18413798-9 2008 The antitumor effect of RGD-L-TRAIL was further enhanced by combination with CPT-11 in both TRAIL-sensitive COLO-205 and TRAIL-resistive HT-29 tumor xenograft models. Irinotecan 77-83 tumor necrosis factor (ligand) superfamily, member 10 Mus musculus 92-97 18413798-9 2008 The antitumor effect of RGD-L-TRAIL was further enhanced by combination with CPT-11 in both TRAIL-sensitive COLO-205 and TRAIL-resistive HT-29 tumor xenograft models. Irinotecan 77-83 tumor necrosis factor (ligand) superfamily, member 10 Mus musculus 92-97 18379174-2 2008 Irinotecan, an anticancer drug, is one of the models for personalized medicine, and a number of clinical studies have revealed significant associations between UGT1A1(*)28 and irinotecan toxicity. Irinotecan 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 160-166 18379174-2 2008 Irinotecan, an anticancer drug, is one of the models for personalized medicine, and a number of clinical studies have revealed significant associations between UGT1A1(*)28 and irinotecan toxicity. Irinotecan 176-186 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 160-166 18379174-6 2008 This mini-review outlines our recent studies on irinotecan pharmacogenetics and discusses the clinical significance of UGT1A1(*)6 and (*)28 markers for personalized irinotecan therapy in Japanese cancer patients. Irinotecan 165-175 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 119-125 18381960-1 2008 PURPOSE: This study was designed to evaluate the in vitro cytotoxicity and in vivo efficacy of TRA-8, a mouse monoclonal antibody that binds to the DR5 death receptor for tumor necrosis factor-related apoptosis-inducing ligand (also called Apo2L), alone and in combination with CPT-11, against human colon cancer cells and xenografts. Irinotecan 278-284 TNF receptor superfamily member 10b Homo sapiens 148-151 18357380-9 2008 The effects of 5-FU and SN-38 on thymidylate synthase (TS) protein expression, an important determinant of 5-FU sensitivity, were assessed by Western blot analysis in H69 cells. Irinotecan 24-29 thymidylate synthetase Homo sapiens 33-53 18357380-9 2008 The effects of 5-FU and SN-38 on thymidylate synthase (TS) protein expression, an important determinant of 5-FU sensitivity, were assessed by Western blot analysis in H69 cells. Irinotecan 24-29 thymidylate synthetase Homo sapiens 55-57 18357380-10 2008 Treatment with SN-38 for 24 h suppressed TS protein expression and this low level of TS was maintained for at least 72 h. Pretreatment with SN-38 inhibited the 5-FU-induced increase of TS protein. Irinotecan 15-20 thymidylate synthetase Homo sapiens 41-43 18357380-10 2008 Treatment with SN-38 for 24 h suppressed TS protein expression and this low level of TS was maintained for at least 72 h. Pretreatment with SN-38 inhibited the 5-FU-induced increase of TS protein. Irinotecan 140-145 thymidylate synthetase Homo sapiens 41-43 18357380-10 2008 Treatment with SN-38 for 24 h suppressed TS protein expression and this low level of TS was maintained for at least 72 h. Pretreatment with SN-38 inhibited the 5-FU-induced increase of TS protein. Irinotecan 140-145 thymidylate synthetase Homo sapiens 85-87 18357380-10 2008 Treatment with SN-38 for 24 h suppressed TS protein expression and this low level of TS was maintained for at least 72 h. Pretreatment with SN-38 inhibited the 5-FU-induced increase of TS protein. Irinotecan 140-145 thymidylate synthetase Homo sapiens 85-87 18357380-11 2008 The synergistic effect induced by the combination of SN-38 and 5-FU may be attributable to the SN-38-induced suppression of TS protein. Irinotecan 53-58 thymidylate synthetase Homo sapiens 124-126 18357380-11 2008 The synergistic effect induced by the combination of SN-38 and 5-FU may be attributable to the SN-38-induced suppression of TS protein. Irinotecan 95-100 thymidylate synthetase Homo sapiens 124-126 18082937-1 2008 Recent pharmacogenetic studies on irinotecan have revealed the impact of UDP glucuronosyltransferase (UGT) 1A1*28 on severe irinotecan toxicities. Irinotecan 34-44 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 73-110 18082937-1 2008 Recent pharmacogenetic studies on irinotecan have revealed the impact of UDP glucuronosyltransferase (UGT) 1A1*28 on severe irinotecan toxicities. Irinotecan 124-134 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 73-110 18082937-2 2008 Although the clinical role of UGT1A1*6, which is specifically detected in East Asian patients, in irinotecan toxicities is suggested, clear evidence remains limited. Irinotecan 98-108 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 30-36 18082937-3 2008 To examine the impact of *6, the association of UGT1A1 genotypes with severe irinotecan toxicities was retrospectively investigated in Japanese cancer patients. Irinotecan 77-87 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 48-54 17483951-1 2008 PURPOSE: Carboxylesterase 2 (CES2) is involved in the activation of the anticancer drug irinotecan to its active metabolite SN-38. Irinotecan 88-98 carboxylesterase 2 Homo sapiens 9-27 18347181-3 2008 CDC45L, NFKB1, PARP1, TDP1, and XRCC1 have been shown to influence the cytotoxic action of camptothecins, including irinotecan. Irinotecan 116-126 cell division cycle 45 Homo sapiens 0-6 18347181-3 2008 CDC45L, NFKB1, PARP1, TDP1, and XRCC1 have been shown to influence the cytotoxic action of camptothecins, including irinotecan. Irinotecan 116-126 nuclear factor kappa B subunit 1 Homo sapiens 8-13 18347181-3 2008 CDC45L, NFKB1, PARP1, TDP1, and XRCC1 have been shown to influence the cytotoxic action of camptothecins, including irinotecan. Irinotecan 116-126 poly(ADP-ribose) polymerase 1 Homo sapiens 15-20 18347181-3 2008 CDC45L, NFKB1, PARP1, TDP1, and XRCC1 have been shown to influence the cytotoxic action of camptothecins, including irinotecan. Irinotecan 116-126 tyrosyl-DNA phosphodiesterase 1 Homo sapiens 22-26 18347181-3 2008 CDC45L, NFKB1, PARP1, TDP1, and XRCC1 have been shown to influence the cytotoxic action of camptothecins, including irinotecan. Irinotecan 116-126 X-ray repair cross complementing 1 Homo sapiens 32-37 17998284-2 2008 PATIENTS AND METHODS: We studied the KRAS mutation status of 113 patients with irinotecan refractory mCRC treated with CTX in clinical trials. Irinotecan 79-89 KRAS proto-oncogene, GTPase Homo sapiens 37-41 17483951-1 2008 PURPOSE: Carboxylesterase 2 (CES2) is involved in the activation of the anticancer drug irinotecan to its active metabolite SN-38. Irinotecan 88-98 carboxylesterase 2 Homo sapiens 29-33 17483951-1 2008 PURPOSE: Carboxylesterase 2 (CES2) is involved in the activation of the anticancer drug irinotecan to its active metabolite SN-38. Irinotecan 124-129 carboxylesterase 2 Homo sapiens 9-27 17483951-1 2008 PURPOSE: Carboxylesterase 2 (CES2) is involved in the activation of the anticancer drug irinotecan to its active metabolite SN-38. Irinotecan 124-129 carboxylesterase 2 Homo sapiens 29-33 17483951-4 2008 CES2 genotypes were tentatively related to irinotecan cytotoxicity and CES2 expression in the NCI-60 panel; to response to treatment and event-free survival in colorectal cancer patients; and to CES2 expression and catalytic activity in subsets of the human liver collection. Irinotecan 43-53 carboxylesterase 2 Homo sapiens 0-4 18349289-3 2008 METHODS: Genotyping of the UGT1A1*28, UGT1A7 N129K/R131K, and UGT1A7-57T/G variants was done in 105 irinotecan-treated patients with metastatic colorectal cancer; adverse events were documented during all 297 treatment cycles and analyzed by Cochran-Mantel-Haenszel, Mann-Whitney, and chi2 tests. Irinotecan 100-110 UDP glucuronosyltransferase family 1 member A7 Homo sapiens 62-68 18188187-4 2008 Therefore, we have developed a mutant human CE (hCE1m6), based on the human liver CE hCE1, that can activate CPT-11 approximately 70-fold more efficiently than the wild-type protein and can be expressed at high levels in mammalian cells. Irinotecan 109-115 carboxylesterase 1 Homo sapiens 48-52 18219662-2 2008 Topoisomerase I inhibitors, including 7-ethyl-10-hydroxycamptothecin (SN-38), are substrates effluxed by BCRP. Irinotecan 38-68 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 105-109 18219662-2 2008 Topoisomerase I inhibitors, including 7-ethyl-10-hydroxycamptothecin (SN-38), are substrates effluxed by BCRP. Irinotecan 70-75 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 105-109 18219662-6 2008 Subsequently, bisulfite sequencing analysis in both PC-6 cells and SN-38-resistant PC-6/SN2-5H, highly expressing BCRP cells was performed to identify the methylated region in the BCRP promoter. Irinotecan 67-72 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 114-118 18219662-6 2008 Subsequently, bisulfite sequencing analysis in both PC-6 cells and SN-38-resistant PC-6/SN2-5H, highly expressing BCRP cells was performed to identify the methylated region in the BCRP promoter. Irinotecan 67-72 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 180-184 18349289-1 2008 BACKGROUND: Gilbert"s syndrome is characterized by a functional promoter single nucleotide polymorphism (SNP) of the UDP-glucuronosyltransferase (UGT) 1A1 gene and represents a pharmacogenetic risk factor for irinotecan toxicity, but study data remain controversial. Irinotecan 209-219 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 117-154 18349289-2 2008 The active CPT-11 metabolite 7-ethyl-10-hydroxycamptothecin is detoxified by several UGT1A proteins, which include UGT1A7 with a high specific activity that may contribute to the risk of irinotecan toxicity in Gilbert"s syndrome patients. Irinotecan 11-17 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 85-90 18349289-7 2008 CONCLUSIONS: Irinotecan toxicity is more likely in patients with Gilbert"s syndrome carrying the UGT1A1*28 allele combined with reduced function UGT1A7 N129K/R131K and UGT1A7-57T/G SNP. Irinotecan 13-23 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 97-103 18349289-2 2008 The active CPT-11 metabolite 7-ethyl-10-hydroxycamptothecin is detoxified by several UGT1A proteins, which include UGT1A7 with a high specific activity that may contribute to the risk of irinotecan toxicity in Gilbert"s syndrome patients. Irinotecan 11-17 UDP glucuronosyltransferase family 1 member A7 Homo sapiens 115-121 18349289-2 2008 The active CPT-11 metabolite 7-ethyl-10-hydroxycamptothecin is detoxified by several UGT1A proteins, which include UGT1A7 with a high specific activity that may contribute to the risk of irinotecan toxicity in Gilbert"s syndrome patients. Irinotecan 29-59 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 85-90 18349289-7 2008 CONCLUSIONS: Irinotecan toxicity is more likely in patients with Gilbert"s syndrome carrying the UGT1A1*28 allele combined with reduced function UGT1A7 N129K/R131K and UGT1A7-57T/G SNP. Irinotecan 13-23 UDP glucuronosyltransferase family 1 member A7 Homo sapiens 145-151 18349289-2 2008 The active CPT-11 metabolite 7-ethyl-10-hydroxycamptothecin is detoxified by several UGT1A proteins, which include UGT1A7 with a high specific activity that may contribute to the risk of irinotecan toxicity in Gilbert"s syndrome patients. Irinotecan 29-59 UDP glucuronosyltransferase family 1 member A7 Homo sapiens 115-121 18349289-7 2008 CONCLUSIONS: Irinotecan toxicity is more likely in patients with Gilbert"s syndrome carrying the UGT1A1*28 allele combined with reduced function UGT1A7 N129K/R131K and UGT1A7-57T/G SNP. Irinotecan 13-23 UDP glucuronosyltransferase family 1 member A7 Homo sapiens 168-174 18349289-2 2008 The active CPT-11 metabolite 7-ethyl-10-hydroxycamptothecin is detoxified by several UGT1A proteins, which include UGT1A7 with a high specific activity that may contribute to the risk of irinotecan toxicity in Gilbert"s syndrome patients. Irinotecan 187-197 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 85-90 18349289-2 2008 The active CPT-11 metabolite 7-ethyl-10-hydroxycamptothecin is detoxified by several UGT1A proteins, which include UGT1A7 with a high specific activity that may contribute to the risk of irinotecan toxicity in Gilbert"s syndrome patients. Irinotecan 187-197 UDP glucuronosyltransferase family 1 member A7 Homo sapiens 115-121 18349289-8 2008 Based on the ability of UGT1A7 to metabolize and eliminate the active irinotecan metabolite 7-ethyl-10-hydroxycamptothecin, the UGT1A1/UGT1A7 SNP combination haplotype appears to be a superior risk predictor than Gilbert"s syndrome alone. Irinotecan 70-80 UDP glucuronosyltransferase family 1 member A7 Homo sapiens 24-30 18349289-8 2008 Based on the ability of UGT1A7 to metabolize and eliminate the active irinotecan metabolite 7-ethyl-10-hydroxycamptothecin, the UGT1A1/UGT1A7 SNP combination haplotype appears to be a superior risk predictor than Gilbert"s syndrome alone. Irinotecan 92-122 UDP glucuronosyltransferase family 1 member A7 Homo sapiens 24-30 18349289-8 2008 Based on the ability of UGT1A7 to metabolize and eliminate the active irinotecan metabolite 7-ethyl-10-hydroxycamptothecin, the UGT1A1/UGT1A7 SNP combination haplotype appears to be a superior risk predictor than Gilbert"s syndrome alone. Irinotecan 92-122 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 128-134 18349289-8 2008 Based on the ability of UGT1A7 to metabolize and eliminate the active irinotecan metabolite 7-ethyl-10-hydroxycamptothecin, the UGT1A1/UGT1A7 SNP combination haplotype appears to be a superior risk predictor than Gilbert"s syndrome alone. Irinotecan 92-122 UDP glucuronosyltransferase family 1 member A7 Homo sapiens 135-141 17786445-8 2008 With irinotecan and 5-FU, mOS around 8 months were reported and with cetuximab combined with irinotecan, the highest mOS was 9.8 months. Irinotecan 5-15 Moloney sarcoma oncogene Mus musculus 26-29 17905465-7 2008 In summary, we showed that a ERCC1-negative protein status was significantly related to tumor responsiveness to neoadjuvant chemotherapy with cisplatin and irinotecan, but such a status was not a clear prognostic predictor to cisplatin-based neoadjuvant therapy in NSCLC patients. Irinotecan 156-166 ERCC excision repair 1, endonuclease non-catalytic subunit Homo sapiens 29-34 18082941-5 2008 Antineoplastic screening demonstrated that five candidates agents, docetaxel, actinomycin D, mitoxantrone, paclitaxel, and SN-38, exhibited potent inhibitory effects on OATP1B3-mediated transport of CDCA-NBD. Irinotecan 123-128 solute carrier organic anion transporter family member 1B3 Homo sapiens 169-176 18082941-7 2008 We determined that SN-38 is a novel substrate for OATP1B3. Irinotecan 19-24 solute carrier organic anion transporter family member 1B3 Homo sapiens 50-57 18023188-1 2008 Carboxylesterases metabolize numerous exogenous and endogenous ester-containing compounds including the chemotherapeutic agent CPT-11, anti-influenza viral agent oseltamivir, and many agrochemicals. Irinotecan 127-133 carboxylesterase 1 Homo sapiens 0-17 18157147-0 2008 Tumor targeting carboxylesterase fused with anti-CEA scFv improve the anticancer effect with a less toxic dose of irinotecan. Irinotecan 114-124 pregnancy specific beta-1-glycoprotein 2 Homo sapiens 49-52 18157147-0 2008 Tumor targeting carboxylesterase fused with anti-CEA scFv improve the anticancer effect with a less toxic dose of irinotecan. Irinotecan 114-124 immunglobulin heavy chain variable region Homo sapiens 53-57 18157147-6 2008 The recombinant enzyme anchors onto the tumor cell surface CEA, and thus metabolize CPT-11 extracellularly. Irinotecan 84-90 pregnancy specific beta-1-glycoprotein 2 Homo sapiens 59-62 18182997-0 2008 Pharmacological targeting of NF-kappaB potentiates the effect of the topoisomerase inhibitor CPT-11 on colon cancer cells. Irinotecan 93-99 nuclear factor kappa B subunit 1 Homo sapiens 29-38 18182997-4 2008 In human colon cancer cells, inhibition of NF-kappaB using 10 microM AS602868 induced a 30-50% growth inhibitory effect and strongly enhanced the action of SN-38, the topoisomerase I inhibitor and CPT-11 active metabolite. Irinotecan 156-161 nuclear factor kappa B subunit 1 Homo sapiens 43-52 18182997-4 2008 In human colon cancer cells, inhibition of NF-kappaB using 10 microM AS602868 induced a 30-50% growth inhibitory effect and strongly enhanced the action of SN-38, the topoisomerase I inhibitor and CPT-11 active metabolite. Irinotecan 197-203 nuclear factor kappa B subunit 1 Homo sapiens 43-52 18182997-6 2008 In xenografts experiments, inhibition of NF-kappaB potentiated the antitumoural effect of CPT-11 in a dose-dependent manner. Irinotecan 90-96 nuclear factor kappa B subunit 1 Homo sapiens 41-50 18182997-8 2008 Ex vivo tumour analyses as well as in vitro studies showed that AS602868 impaired CPT-11-induced NF-kappaB activation, and enhanced tumour cell cycle arrest and apoptosis. Irinotecan 82-88 nuclear factor kappa B subunit 1 Homo sapiens 97-106 18182667-2 2008 The aim of these retrospective studies was to evaluate tumor vascularity and expression of components of the VEGF pathway and hypoxic responses as predictive markers for radiographic response and survival benefit from the bevacizumab and irinotecan therapy. Irinotecan 238-248 vascular endothelial growth factor A Homo sapiens 109-113 18230579-2 2008 This chimeric antibody inhibiting the Epidermal Growth receptor (EGFR) has been demonstrated to be efficient in the treatment of irinotecan-resistant metastatic CRC expressing the EGFR. Irinotecan 129-139 epidermal growth factor receptor Homo sapiens 65-69 18230579-2 2008 This chimeric antibody inhibiting the Epidermal Growth receptor (EGFR) has been demonstrated to be efficient in the treatment of irinotecan-resistant metastatic CRC expressing the EGFR. Irinotecan 129-139 epidermal growth factor receptor Homo sapiens 180-184 18231104-5 2008 In patients given first-line cisplatin-based regimens (combined with S-1 or irinotecan) (43 patients), low ERCC1 correlated with a higher response rate (low: 55.6% vs high: 18.8%; P=0.008). Irinotecan 76-86 ERCC excision repair 1, endonuclease non-catalytic subunit Homo sapiens 107-112 18281753-1 2008 CPT-11 is activated by hydrolysis to SN-38 that is primarily inactivated through biotrans-formation into SN-38 glucuronide ( SN-38 G) by UGT1As. Irinotecan 0-6 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 137-141 18281753-1 2008 CPT-11 is activated by hydrolysis to SN-38 that is primarily inactivated through biotrans-formation into SN-38 glucuronide ( SN-38 G) by UGT1As. Irinotecan 37-42 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 137-141 18281753-1 2008 CPT-11 is activated by hydrolysis to SN-38 that is primarily inactivated through biotrans-formation into SN-38 glucuronide ( SN-38 G) by UGT1As. Irinotecan 105-110 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 137-141 18281753-3 2008 The toxicities of CPT-11 have been reported to correlate with the polymorphism of the number of TA repeats in UGT1A1 gene because of its gene transcriptional efficiency. Irinotecan 18-24 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 110-116 18281776-6 2008 She was also treated with cisplatin and S-1 as fourth-line chemotherapy after treatment with nedaplatin and gemcitabine, docetaxel and irinotecan, and gefitinib. Irinotecan 135-145 proteasome 26S subunit, non-ATPase 1 Homo sapiens 40-43 18778115-1 2008 Bevacizumab (Avastin) is a recombinant, humanized monoclonal antibody against vascular endothelial growth factor (VEGF) that is used to inhibit VEGF function in vascular endothelial cells and thereby inhibit tumor angiogenesis, upon which solid tumors depend for growth and metastasis.The addition of bevacizumab to fluoropyrimidine-based chemotherapy, with or without irinotecan or oxaliplatin, in both the first- and second-line treatment of metastatic colorectal cancer, significantly increased median progression-free survival or time to disease progression in most randomized controlled trials. Irinotecan 369-379 vascular endothelial growth factor A Homo sapiens 78-112 18778115-1 2008 Bevacizumab (Avastin) is a recombinant, humanized monoclonal antibody against vascular endothelial growth factor (VEGF) that is used to inhibit VEGF function in vascular endothelial cells and thereby inhibit tumor angiogenesis, upon which solid tumors depend for growth and metastasis.The addition of bevacizumab to fluoropyrimidine-based chemotherapy, with or without irinotecan or oxaliplatin, in both the first- and second-line treatment of metastatic colorectal cancer, significantly increased median progression-free survival or time to disease progression in most randomized controlled trials. Irinotecan 369-379 vascular endothelial growth factor A Homo sapiens 114-118 19075668-13 2008 Using an ex vivo drug sensitivity assay, the histoculture drug response assay (HDRA), the expression of ATP7A in human surgically resected colon cancer cells correlated with sensitivity to 7-ethyl-10-hydroxy-camptothecin (SN-38). Irinotecan 222-227 ATPase copper transporting alpha Homo sapiens 104-109 19075668-14 2008 ATP7A-overexpressing cells are resistant to many anticancer drugs including SN-38, 7-ethyl-10-[4-(1-piperidino)-1-piperidino] carbonyloxycamptothecin (CPT-11), vincristine, paclitaxel, etoposide, doxorubicin (Dox), and mitoxantron. Irinotecan 76-81 ATPase copper transporting alpha Homo sapiens 0-5 19075668-14 2008 ATP7A-overexpressing cells are resistant to many anticancer drugs including SN-38, 7-ethyl-10-[4-(1-piperidino)-1-piperidino] carbonyloxycamptothecin (CPT-11), vincristine, paclitaxel, etoposide, doxorubicin (Dox), and mitoxantron. Irinotecan 83-149 ATPase copper transporting alpha Homo sapiens 0-5 19075668-14 2008 ATP7A-overexpressing cells are resistant to many anticancer drugs including SN-38, 7-ethyl-10-[4-(1-piperidino)-1-piperidino] carbonyloxycamptothecin (CPT-11), vincristine, paclitaxel, etoposide, doxorubicin (Dox), and mitoxantron. Irinotecan 151-157 ATPase copper transporting alpha Homo sapiens 0-5 17786445-8 2008 With irinotecan and 5-FU, mOS around 8 months were reported and with cetuximab combined with irinotecan, the highest mOS was 9.8 months. Irinotecan 93-103 Moloney sarcoma oncogene Mus musculus 117-120 17766002-0 2008 Influence of the organic anion-transporting polypeptide 1B1 (OATP1B1) polymorphisms on irinotecan-pharmacokinetics and clinical outcome of patients with advanced non-small cell lung cancer. Irinotecan 87-97 solute carrier organic anion transporter family member 1B1 Homo sapiens 17-59 18370855-1 2008 ABCG2 was discovered in multi-drug-resistant cancer cells, with the identification of chemotherapeutic agents, such as mitoxantrone, flavopiridol, methotrexate and irinotecan as substrates. Irinotecan 164-174 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 0-5 18714167-7 2008 CONCLUSION: These results indicate that both Apo2L/TRAIL-sensitive and -resistant colon tumors will respond to a combination of CPT-11 and Apo2L/TRAIL and predict that this will be useful in the treatment of human colon cancers in a clinical setting. Irinotecan 128-134 TNF superfamily member 10 Homo sapiens 45-50 18714167-7 2008 CONCLUSION: These results indicate that both Apo2L/TRAIL-sensitive and -resistant colon tumors will respond to a combination of CPT-11 and Apo2L/TRAIL and predict that this will be useful in the treatment of human colon cancers in a clinical setting. Irinotecan 128-134 TNF superfamily member 10 Homo sapiens 51-56 17766002-0 2008 Influence of the organic anion-transporting polypeptide 1B1 (OATP1B1) polymorphisms on irinotecan-pharmacokinetics and clinical outcome of patients with advanced non-small cell lung cancer. Irinotecan 87-97 solute carrier organic anion transporter family member 1B1 Homo sapiens 61-68 18852492-3 2008 This model was applied to data from phase II/III trials of first-line bevacizumab plus 5-FU/LV with/without irinotecan (IFL). Irinotecan 108-118 interferon alpha 1 Homo sapiens 120-123 17766002-1 2008 To investigate whether the OATP1B1 polymorphisms affect irinotecan-pharmacokinetics and subsequent toxicity and tumor response of patients with advanced NSCLC. Irinotecan 56-66 solute carrier organic anion transporter family member 1B1 Homo sapiens 27-34 17766002-8 2008 These findings suggest that OATP1B1 variants are involved in SN-38 disposition and highly predictive for severe toxicity of NSCLC patients treated with irinotecan-based chemotherapy. Irinotecan 61-66 solute carrier organic anion transporter family member 1B1 Homo sapiens 28-35 17766002-8 2008 These findings suggest that OATP1B1 variants are involved in SN-38 disposition and highly predictive for severe toxicity of NSCLC patients treated with irinotecan-based chemotherapy. Irinotecan 152-162 solute carrier organic anion transporter family member 1B1 Homo sapiens 28-35 17345086-9 2007 Among these active compounds, 3",4",7-trimethoxyflavone showed the strongest anti-BCRP activity with RI(50) values of 0.012 microM for SN-38 and 0.044 muM for mitoxantrone. Irinotecan 135-140 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 82-86 17898154-1 2007 The UGT1A1*28 polymorphism is known to correlate with altered clearance of bilirubin (Gilbert syndrome) and drugs such as 7-ethyl-10-[4-(1-piperidino)-1-piperidino] carbonyloxy camptothecin (CPT-11). Irinotecan 122-189 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 17898154-1 2007 The UGT1A1*28 polymorphism is known to correlate with altered clearance of bilirubin (Gilbert syndrome) and drugs such as 7-ethyl-10-[4-(1-piperidino)-1-piperidino] carbonyloxy camptothecin (CPT-11). Irinotecan 191-197 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 17898154-5 2007 Bilirubin, estradiol (3-OH), ethinyl estradiol (3-OH), and 7-ethyl-10-hydroxycamptothecin (SN-38) were found to show significantly lower rates of metabolism in the UGT1A1*28/*28 microsomes with no change in K(m) values. Irinotecan 59-89 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 164-170 17978195-4 2007 Strain-specific differences in the rate of 16 alpha-hydroxytestosterone generation and irinotecan glucuronidation correlated with the pattern of genetic variation within Cyp2b9 and Ugt1a loci, respectively. Irinotecan 87-97 cytochrome P450, family 2, subfamily b, polypeptide 9 Mus musculus 170-176 17978195-4 2007 Strain-specific differences in the rate of 16 alpha-hydroxytestosterone generation and irinotecan glucuronidation correlated with the pattern of genetic variation within Cyp2b9 and Ugt1a loci, respectively. Irinotecan 87-97 Ugt1a@ Mus musculus 181-186 18361806-3 2007 The anti-EGFR monoclonal antibody cetuximab has been shown to reverse chemotherapy resistance in patients with irinotecan-refractory mCRC, to improve survival compared with best supportive care (BSC) alone, and to prolong progression free-survival (PFS) in the first- and second-line settings. Irinotecan 111-121 epidermal growth factor receptor Homo sapiens 9-13 18478930-1 2007 OBJECTIVE: To assess the polymorphism of UGT1A gene in Chinese, and to investigate the correlation between UGT1A polymorphism and irinotecan toxicity in colorectal cancer patients. Irinotecan 130-140 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 107-112 18478930-8 2007 CONCLUSION: Chinese patients exhibit less irinotecan-related diarrhea due to higher frequence of UGT1A A1 * 28 wild genotype TA6/6. Irinotecan 42-52 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 97-102 17273826-5 2007 A decrease in Bcl-xL sensitised cells to the small molecule inhibitor of Bcl-xL, Antimycin A3 and the DNA topoisomerase I inhibitors, SN-38 and camptothecin, but not to doxorubicin. Irinotecan 134-139 BCL2 like 1 Homo sapiens 14-20 29793274-2 2007 : Role ofUGT1A1*6, UGT1A1*28 and ABCG2 c.421C>A polymorphisms in irinotecan-induced neutropenia in Asian cancer patients. Irinotecan 68-78 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 33-38 29793274-7 2007 In that study, a statistically significantly higher level of SN-38 and a relatively lower degree of glucuronidation occurred in patients with the UGT1A1*6 homozygote genotype than in patients with the reference genotype. Irinotecan 61-66 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 146-152 17942911-9 2007 Knockdown of GRP78 also sensitizes glioma cells to 5-fluorouracil and CPT-11. Irinotecan 70-76 heat shock protein family A (Hsp70) member 5 Homo sapiens 13-18 17273826-0 2007 Short hairpin RNAs targeting Bcl-xL modulate senescence and apoptosis following SN-38 and irinotecan exposure in a colon cancer model. Irinotecan 80-85 BCL2 like 1 Homo sapiens 29-35 17273826-0 2007 Short hairpin RNAs targeting Bcl-xL modulate senescence and apoptosis following SN-38 and irinotecan exposure in a colon cancer model. Irinotecan 90-100 BCL2 like 1 Homo sapiens 29-35 17273826-10 2007 Antimycin A3, in combination with SN-38 recapitulated this phenotype in HCT 116 cells, suggesting a potential role for small molecule inhibitors of Bcl-xL/Bcl-2 in the treatment of colorectal cancer, potentially in combination with irinotecan. Irinotecan 34-39 BCL2 like 1 Homo sapiens 148-154 17728214-7 2007 The risk of experiencing irinotecan-induced hematologic toxicity for patients with a UGT1A1*28/*28 genotype thus appears to be a function of the dose of irinotecan administered. Irinotecan 25-35 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 85-91 17393318-11 2007 MEK activation suppressed CPT-11-induced apoptosis in IEC-caMEK cells via a COX-2- dependent mechanism. Irinotecan 26-32 cytochrome c oxidase II, mitochondrial Rattus norvegicus 76-81 17898662-0 2007 Severe toxicities after irinotecan-based chemotherapy in a patient with lung cancer: a homozygote for the SLCO1B1*15 allele. Irinotecan 24-34 solute carrier organic anion transporter family member 1B1 Homo sapiens 106-113 17898662-2 2007 Polymorphism of the UGT1A1 gene is one of the likely reasons for interindividual differences in irinotecan pharmacokinetics and severe toxicity. Irinotecan 96-106 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 20-26 17898662-3 2007 Also, polymorphic organic anion-transporting polypeptide 1B1 (OATP1B1, SLCO1B1) is reported to be involved in the hepatocellular uptake of SN-38. Irinotecan 139-144 solute carrier organic anion transporter family member 1B1 Homo sapiens 18-60 17898662-3 2007 Also, polymorphic organic anion-transporting polypeptide 1B1 (OATP1B1, SLCO1B1) is reported to be involved in the hepatocellular uptake of SN-38. Irinotecan 139-144 solute carrier organic anion transporter family member 1B1 Homo sapiens 62-69 17898662-3 2007 Also, polymorphic organic anion-transporting polypeptide 1B1 (OATP1B1, SLCO1B1) is reported to be involved in the hepatocellular uptake of SN-38. Irinotecan 139-144 solute carrier organic anion transporter family member 1B1 Homo sapiens 71-78 17898662-6 2007 Analysis of genetic variants in genes encoding the drug-metabolizing enzyme (UGT1A1) and transporter (SLCO1B1) involving irinotecan disposition revealed that this patient was homozygous for the SLCO1B1*15 allele, which may result in severe toxicities attributable to the extensive accumulation of SN-38. Irinotecan 121-131 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 77-83 17898662-6 2007 Analysis of genetic variants in genes encoding the drug-metabolizing enzyme (UGT1A1) and transporter (SLCO1B1) involving irinotecan disposition revealed that this patient was homozygous for the SLCO1B1*15 allele, which may result in severe toxicities attributable to the extensive accumulation of SN-38. Irinotecan 121-131 solute carrier organic anion transporter family member 1B1 Homo sapiens 102-109 17898662-6 2007 Analysis of genetic variants in genes encoding the drug-metabolizing enzyme (UGT1A1) and transporter (SLCO1B1) involving irinotecan disposition revealed that this patient was homozygous for the SLCO1B1*15 allele, which may result in severe toxicities attributable to the extensive accumulation of SN-38. Irinotecan 121-131 solute carrier organic anion transporter family member 1B1 Homo sapiens 194-201 17898662-6 2007 Analysis of genetic variants in genes encoding the drug-metabolizing enzyme (UGT1A1) and transporter (SLCO1B1) involving irinotecan disposition revealed that this patient was homozygous for the SLCO1B1*15 allele, which may result in severe toxicities attributable to the extensive accumulation of SN-38. Irinotecan 297-302 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 77-83 17898662-6 2007 Analysis of genetic variants in genes encoding the drug-metabolizing enzyme (UGT1A1) and transporter (SLCO1B1) involving irinotecan disposition revealed that this patient was homozygous for the SLCO1B1*15 allele, which may result in severe toxicities attributable to the extensive accumulation of SN-38. Irinotecan 297-302 solute carrier organic anion transporter family member 1B1 Homo sapiens 102-109 17898662-6 2007 Analysis of genetic variants in genes encoding the drug-metabolizing enzyme (UGT1A1) and transporter (SLCO1B1) involving irinotecan disposition revealed that this patient was homozygous for the SLCO1B1*15 allele, which may result in severe toxicities attributable to the extensive accumulation of SN-38. Irinotecan 297-302 solute carrier organic anion transporter family member 1B1 Homo sapiens 194-201 17898662-7 2007 Screening of SLCO1B1*15 is suggested to be useful in irinotecan chemotherapy to avoid unpredicted severe toxicity, although the homozygous genotype is rare among the Japanese. Irinotecan 53-63 solute carrier organic anion transporter family member 1B1 Homo sapiens 13-20 17728214-1 2007 The Food and Drug Administration and Pfizer changed the package insert for irinotecan to include a patient"s UGT1A1*28 genotype as a risk factor for severe neutropenia on the basis of the findings of four pharmacogenetic studies, which found that irinotecan-treated patients who were homozygous for the UGT1A1*28 allele had a greater risk of hematologic toxic effects than patients who had one or two copies of the wild-type allele (UGT1A1*1). Irinotecan 75-85 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 109-115 17728214-1 2007 The Food and Drug Administration and Pfizer changed the package insert for irinotecan to include a patient"s UGT1A1*28 genotype as a risk factor for severe neutropenia on the basis of the findings of four pharmacogenetic studies, which found that irinotecan-treated patients who were homozygous for the UGT1A1*28 allele had a greater risk of hematologic toxic effects than patients who had one or two copies of the wild-type allele (UGT1A1*1). Irinotecan 247-257 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 109-115 17640957-2 2007 hCE-2 also activates an anticancer drug, irinotecan (7-ethyl-10-[4-(1-piperidino)-1-piperidino]-carbonyloxycamptothecin, CPT-11), into its active metabolite, 7-ethyl-10-hydroxycamptothecin (SN-38). Irinotecan 41-51 carboxylesterase 2 Homo sapiens 0-5 17640957-2 2007 hCE-2 also activates an anticancer drug, irinotecan (7-ethyl-10-[4-(1-piperidino)-1-piperidino]-carbonyloxycamptothecin, CPT-11), into its active metabolite, 7-ethyl-10-hydroxycamptothecin (SN-38). Irinotecan 53-119 carboxylesterase 2 Homo sapiens 0-5 17640957-2 2007 hCE-2 also activates an anticancer drug, irinotecan (7-ethyl-10-[4-(1-piperidino)-1-piperidino]-carbonyloxycamptothecin, CPT-11), into its active metabolite, 7-ethyl-10-hydroxycamptothecin (SN-38). Irinotecan 121-127 carboxylesterase 2 Homo sapiens 0-5 17640957-2 2007 hCE-2 also activates an anticancer drug, irinotecan (7-ethyl-10-[4-(1-piperidino)-1-piperidino]-carbonyloxycamptothecin, CPT-11), into its active metabolite, 7-ethyl-10-hydroxycamptothecin (SN-38). Irinotecan 158-188 carboxylesterase 2 Homo sapiens 0-5 17640957-2 2007 hCE-2 also activates an anticancer drug, irinotecan (7-ethyl-10-[4-(1-piperidino)-1-piperidino]-carbonyloxycamptothecin, CPT-11), into its active metabolite, 7-ethyl-10-hydroxycamptothecin (SN-38). Irinotecan 190-195 carboxylesterase 2 Homo sapiens 0-5 17060921-1 2007 Variants within the human UGT1A1 gene are associated with irinotecan induced severely adverse reactions and hyperbilirubinemia. Irinotecan 58-68 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 26-32 17875772-11 2007 Both in vitro and in vivo studies suggest inactivation of MGMT with O(6)-benzylguanine may increase the activity of temozolomide and irinotecan against neuroblastoma. Irinotecan 133-143 O-6-methylguanine-DNA methyltransferase Mus musculus 58-62 17728214-7 2007 The risk of experiencing irinotecan-induced hematologic toxicity for patients with a UGT1A1*28/*28 genotype thus appears to be a function of the dose of irinotecan administered. Irinotecan 153-163 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 85-91 19262706-1 2007 PURPOSE: To evaluate the association between dihydropyrimidine dehydrogenase (DPD) and thymidylate synthase (TS) levels in primary gastric tumors and clinical response to S-1 or S-1 plus irinotecan in patients with unresectable advanced gastric cancer, and to investigate the molecular mechanism of augmented antitumor activity of the combination using human gastric cancer xenografts with high TS activity. Irinotecan 187-197 dihydropyrimidine dehydrogenase Homo sapiens 45-76 17627617-7 2007 These exploratory findings suggest that homozygosity for UGT1A1*6 allele may be associated with altered SN-38 disposition and may increase the risk of severe neutropenia in Asian cancer patients, particularly in the Chinese cancer patients who comprised 80% (n = 36) of the patient population in the present study. Irinotecan 104-109 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 57-63 17406868-0 2007 Genetic linkage of UGT1A7 and UGT1A9 polymorphisms to UGT1A1*6 is associated with reduced activity for SN-38 in Japanese patients with cancer. Irinotecan 103-108 UDP glucuronosyltransferase family 1 member A7 Homo sapiens 19-25 17406868-0 2007 Genetic linkage of UGT1A7 and UGT1A9 polymorphisms to UGT1A1*6 is associated with reduced activity for SN-38 in Japanese patients with cancer. Irinotecan 103-108 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 30-36 17406868-0 2007 Genetic linkage of UGT1A7 and UGT1A9 polymorphisms to UGT1A1*6 is associated with reduced activity for SN-38 in Japanese patients with cancer. Irinotecan 103-108 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 54-60 17406868-1 2007 PURPOSE: The phenotypic effects of UGT1A7 and UGT1A9 genetic polymorphisms on the in vivo pharmacokinetics of irinotecan were examined. Irinotecan 110-120 UDP glucuronosyltransferase family 1 member A7 Homo sapiens 35-41 17406868-1 2007 PURPOSE: The phenotypic effects of UGT1A7 and UGT1A9 genetic polymorphisms on the in vivo pharmacokinetics of irinotecan were examined. Irinotecan 110-120 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 46-52 17406868-9 2007 CONCLUSION: Genetic linkage of UGT1A7 and UGT1A9 polymorphisms to UGT1A1*6, related to reduced catalytic and transcriptional activities of UGTs, is associated with the decreased glucuronosyltransferase activity for SN-38 in Japanese patients with cancer. Irinotecan 215-220 UDP glucuronosyltransferase family 1 member A7 Homo sapiens 31-37 17406868-9 2007 CONCLUSION: Genetic linkage of UGT1A7 and UGT1A9 polymorphisms to UGT1A1*6, related to reduced catalytic and transcriptional activities of UGTs, is associated with the decreased glucuronosyltransferase activity for SN-38 in Japanese patients with cancer. Irinotecan 215-220 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 42-48 17406868-9 2007 CONCLUSION: Genetic linkage of UGT1A7 and UGT1A9 polymorphisms to UGT1A1*6, related to reduced catalytic and transcriptional activities of UGTs, is associated with the decreased glucuronosyltransferase activity for SN-38 in Japanese patients with cancer. Irinotecan 215-220 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 66-72 17873515-1 2007 PURPOSE: Because of its supposed inhibiting capacities of uridine-diphosphate glucuronosyltransferase 1A (UGT1A)-mediated glucuronidation in rats, the antiepileptic drug valproic acid has been investigated as modulator of irinotecan-induced delayed-type diarrhea in rats. Irinotecan 222-232 Ugt1a@ Rattus norvegicus 106-111 19262706-1 2007 PURPOSE: To evaluate the association between dihydropyrimidine dehydrogenase (DPD) and thymidylate synthase (TS) levels in primary gastric tumors and clinical response to S-1 or S-1 plus irinotecan in patients with unresectable advanced gastric cancer, and to investigate the molecular mechanism of augmented antitumor activity of the combination using human gastric cancer xenografts with high TS activity. Irinotecan 187-197 dihydropyrimidine dehydrogenase Homo sapiens 78-81 19262706-5 2007 In the experimental studies, S-1 plus irinotecan showed augmented antitumor activity against tumors with high TS activity (P < .01) compared with either agent alone. Irinotecan 38-48 thymidylate synthetase Homo sapiens 110-112 19262706-6 2007 A potential mechanism for this effect was suggested by the significant reduction in TS activity observed following irinotecan administration in tumors with high TS activity. Irinotecan 115-125 thymidylate synthetase Homo sapiens 84-86 19262706-6 2007 A potential mechanism for this effect was suggested by the significant reduction in TS activity observed following irinotecan administration in tumors with high TS activity. Irinotecan 115-125 thymidylate synthetase Homo sapiens 161-163 19262706-7 2007 CONCLUSION: This study suggests that, via down-regulation of TS by irinotecan treatment, combination chemotherapy with S-1 and irinotecan could be effective in gastric cancer patients with high TS levels. Irinotecan 67-77 thymidylate synthetase Homo sapiens 61-63 19262706-7 2007 CONCLUSION: This study suggests that, via down-regulation of TS by irinotecan treatment, combination chemotherapy with S-1 and irinotecan could be effective in gastric cancer patients with high TS levels. Irinotecan 67-77 thymidylate synthetase Homo sapiens 194-196 19262706-7 2007 CONCLUSION: This study suggests that, via down-regulation of TS by irinotecan treatment, combination chemotherapy with S-1 and irinotecan could be effective in gastric cancer patients with high TS levels. Irinotecan 127-137 thymidylate synthetase Homo sapiens 61-63 19262706-7 2007 CONCLUSION: This study suggests that, via down-regulation of TS by irinotecan treatment, combination chemotherapy with S-1 and irinotecan could be effective in gastric cancer patients with high TS levels. Irinotecan 127-137 thymidylate synthetase Homo sapiens 194-196 17634538-0 2007 TIMP-1 is significantly associated with objective response and survival in metastatic colorectal cancer patients receiving combination of irinotecan, 5-fluorouracil, and folinic acid. Irinotecan 138-148 TIMP metallopeptidase inhibitor 1 Homo sapiens 0-6 17691917-5 2007 So far, the candidate gene approach has provided important clues for pharmacogenomic-based personalized chemotherapy with 6-mercaptopurine (6-MP), solely metabolized by thiopurine S-methyltransferase (TPMT), and irinotecan, mainly detoxified by UDP-glucuronosyltransferase 1A1 (UGT1A1). Irinotecan 212-222 thiopurine S-methyltransferase Homo sapiens 201-205 17691917-6 2007 Reduced activity of TPMT caused by polymorphisms in the TPMT gene and decreased activity of UGT1A1 caused by UGT1A1*28 are related to severe toxic effects of 6-MP and irinotecan, respectively. Irinotecan 167-177 thiopurine S-methyltransferase Homo sapiens 20-24 17691917-6 2007 Reduced activity of TPMT caused by polymorphisms in the TPMT gene and decreased activity of UGT1A1 caused by UGT1A1*28 are related to severe toxic effects of 6-MP and irinotecan, respectively. Irinotecan 167-177 thiopurine S-methyltransferase Homo sapiens 56-60 17691917-6 2007 Reduced activity of TPMT caused by polymorphisms in the TPMT gene and decreased activity of UGT1A1 caused by UGT1A1*28 are related to severe toxic effects of 6-MP and irinotecan, respectively. Irinotecan 167-177 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 92-98 17691917-6 2007 Reduced activity of TPMT caused by polymorphisms in the TPMT gene and decreased activity of UGT1A1 caused by UGT1A1*28 are related to severe toxic effects of 6-MP and irinotecan, respectively. Irinotecan 167-177 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 109-115 17537833-0 2007 Interleukin-6 alters the cellular responsiveness to clopidogrel, irinotecan, and oseltamivir by suppressing the expression of carboxylesterases HCE1 and HCE2. Irinotecan 65-75 interleukin 6 Homo sapiens 0-13 17537833-0 2007 Interleukin-6 alters the cellular responsiveness to clopidogrel, irinotecan, and oseltamivir by suppressing the expression of carboxylesterases HCE1 and HCE2. Irinotecan 65-75 carboxylesterase 2 Homo sapiens 126-143 17537833-0 2007 Interleukin-6 alters the cellular responsiveness to clopidogrel, irinotecan, and oseltamivir by suppressing the expression of carboxylesterases HCE1 and HCE2. Irinotecan 65-75 RNA guanylyltransferase and 5'-phosphatase Homo sapiens 144-148 17537833-0 2007 Interleukin-6 alters the cellular responsiveness to clopidogrel, irinotecan, and oseltamivir by suppressing the expression of carboxylesterases HCE1 and HCE2. Irinotecan 65-75 carboxylesterase 2 Homo sapiens 153-157 17666793-0 2007 Irinotecan-induced apoptosis is inhibited by increased P-glycoprotein expression and decreased p53 in human hepatocellular carcinoma cells. Irinotecan 0-10 ATP binding cassette subfamily B member 1 Homo sapiens 55-69 17666793-0 2007 Irinotecan-induced apoptosis is inhibited by increased P-glycoprotein expression and decreased p53 in human hepatocellular carcinoma cells. Irinotecan 0-10 tumor protein p53 Homo sapiens 95-98 17666793-7 2007 SN-38 significantly induced apoptosis in Huh7 cells at 24 h. SN-38 also increased the expression of p53, Bax, and caspase-9 and decreased Bcl-xL expression in Huh7 cells. Irinotecan 0-5 tumor protein p53 Homo sapiens 100-103 17666793-7 2007 SN-38 significantly induced apoptosis in Huh7 cells at 24 h. SN-38 also increased the expression of p53, Bax, and caspase-9 and decreased Bcl-xL expression in Huh7 cells. Irinotecan 0-5 BCL2 associated X, apoptosis regulator Homo sapiens 105-108 17666793-7 2007 SN-38 significantly induced apoptosis in Huh7 cells at 24 h. SN-38 also increased the expression of p53, Bax, and caspase-9 and decreased Bcl-xL expression in Huh7 cells. Irinotecan 0-5 caspase 9 Homo sapiens 114-123 17666793-7 2007 SN-38 significantly induced apoptosis in Huh7 cells at 24 h. SN-38 also increased the expression of p53, Bax, and caspase-9 and decreased Bcl-xL expression in Huh7 cells. Irinotecan 0-5 BCL2 like 1 Homo sapiens 138-144 17666793-7 2007 SN-38 significantly induced apoptosis in Huh7 cells at 24 h. SN-38 also increased the expression of p53, Bax, and caspase-9 and decreased Bcl-xL expression in Huh7 cells. Irinotecan 61-66 tumor protein p53 Homo sapiens 100-103 17666793-7 2007 SN-38 significantly induced apoptosis in Huh7 cells at 24 h. SN-38 also increased the expression of p53, Bax, and caspase-9 and decreased Bcl-xL expression in Huh7 cells. Irinotecan 61-66 BCL2 associated X, apoptosis regulator Homo sapiens 105-108 17666793-7 2007 SN-38 significantly induced apoptosis in Huh7 cells at 24 h. SN-38 also increased the expression of p53, Bax, and caspase-9 and decreased Bcl-xL expression in Huh7 cells. Irinotecan 61-66 caspase 9 Homo sapiens 114-123 17666793-7 2007 SN-38 significantly induced apoptosis in Huh7 cells at 24 h. SN-38 also increased the expression of p53, Bax, and caspase-9 and decreased Bcl-xL expression in Huh7 cells. Irinotecan 61-66 BCL2 like 1 Homo sapiens 138-144 17666793-8 2007 SN-38 decreased p53 expression and increased P-gp expression after 120 h, resulting in inhibition of apoptosis. Irinotecan 0-5 tumor protein p53 Homo sapiens 16-19 17666793-8 2007 SN-38 decreased p53 expression and increased P-gp expression after 120 h, resulting in inhibition of apoptosis. Irinotecan 0-5 ATP binding cassette subfamily B member 1 Homo sapiens 45-49 17666793-10 2007 SN-38-induced P-gp expression was additionally enhanced by p53 decoy oligodeoxynucleotide. Irinotecan 0-5 ATP binding cassette subfamily B member 1 Homo sapiens 14-18 17666793-10 2007 SN-38-induced P-gp expression was additionally enhanced by p53 decoy oligodeoxynucleotide. Irinotecan 0-5 tumor protein p53 Homo sapiens 59-62 17622938-6 2007 RESULTS: A strong correlation was observed between the glucuronidation of T4 and SN-38, a UGT1A1 substrate (r=0.82, P<0.0001). Irinotecan 81-86 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 90-96 17762398-2 2007 The presence of an additional TA repeat in the TATA sequence of the UGT1A1 promoter leads to a significant decrease in SN-38 glucuronidation. Irinotecan 119-124 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 68-74 17762398-3 2007 Patients with the UGT1A1 (TA)7 allele are more likely to experience severe neutropenia and diarrhea following irinotecan chemotherapy. Irinotecan 110-120 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 18-24 17577039-0 2007 UGT1A1 promoter genotype correlates with SN-38 pharmacokinetics, but not severe toxicity in patients receiving low-dose irinotecan. Irinotecan 41-46 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 17534875-0 2007 Associations of ABCB1, ABCC2, and ABCG2 polymorphisms with irinotecan-pharmacokinetics and clinical outcome in patients with advanced non-small cell lung cancer. Irinotecan 59-69 ATP binding cassette subfamily B member 1 Homo sapiens 16-21 17534875-0 2007 Associations of ABCB1, ABCC2, and ABCG2 polymorphisms with irinotecan-pharmacokinetics and clinical outcome in patients with advanced non-small cell lung cancer. Irinotecan 59-69 ATP binding cassette subfamily C member 2 Homo sapiens 23-28 17534875-0 2007 Associations of ABCB1, ABCC2, and ABCG2 polymorphisms with irinotecan-pharmacokinetics and clinical outcome in patients with advanced non-small cell lung cancer. Irinotecan 59-69 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 34-39 17534875-1 2007 BACKGROUND: The authors investigated whether ABCB1, ABCC2, and ABCG2 genetic polymorphisms affect pharmacokinetics (PK) of irinotecan and treatment outcome of patients with advanced nonsmall cell lung cancer (NSCLC). Irinotecan 123-133 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 63-68 17534875-11 2007 CONCLUSIONS: Specific polymorphisms of ABCB1 and ABCC2 can influence disposition and tumor response to irinotecan by regulating transporter activity. Irinotecan 103-113 ATP binding cassette subfamily B member 1 Homo sapiens 39-44 17534875-11 2007 CONCLUSIONS: Specific polymorphisms of ABCB1 and ABCC2 can influence disposition and tumor response to irinotecan by regulating transporter activity. Irinotecan 103-113 ATP binding cassette subfamily C member 2 Homo sapiens 49-54 17681105-1 2007 BACKGROUND: Uridine diphosphate glucuronosyltransferase (UGT) 1A1 7/7 polymorphism has been linked with an increased risk of irinotecan-induced severe toxicities. Irinotecan 125-135 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-65 17681105-2 2007 We evaluated UGT1A1 polymorphism in patients developing grade 3/4 toxicity after initiation of irinotecan to determine the frequency of this polymorphism in this population. Irinotecan 95-105 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 13-19 17681105-3 2007 PATIENTS AND METHODS: Twenty patients with grade 3/4 irinotecan-induced toxicity underwent UGT1A1 genotyping in an exploratory study. Irinotecan 53-63 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 91-97 17681105-8 2007 Our data support the need for more prospective studies that evaluate the predictive value of UGT1A1 as well as UGT1A1-based dosing in patients receiving irinotecan. Irinotecan 153-163 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-99 17681105-8 2007 Our data support the need for more prospective studies that evaluate the predictive value of UGT1A1 as well as UGT1A1-based dosing in patients receiving irinotecan. Irinotecan 153-163 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 111-117 17609585-6 2007 SN-38 binding motifs were detected in the proximal promoter of p53 (bases -433 to -317 and -814 to -711). Irinotecan 0-5 tumor protein p53 Homo sapiens 63-66 17609585-7 2007 These results suggest that the p53-mediated apoptosis pathway is important in the anticancer effects of irinotecan in hepatocellular carcinoma. Irinotecan 104-114 tumor protein p53 Homo sapiens 31-34 17558305-1 2007 OBJECTIVES: SN-38, an active metabolite of irinotecan, is detoxified by glucuronidation with UGT1A isoforms, 1A1, 1A7, 1A9, and 1A10. Irinotecan 12-17 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 93-98 17558305-1 2007 OBJECTIVES: SN-38, an active metabolite of irinotecan, is detoxified by glucuronidation with UGT1A isoforms, 1A1, 1A7, 1A9, and 1A10. Irinotecan 43-53 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 93-98 17558305-2 2007 The pharmacogenetic information on UGT1A haplotypes covering all these isoforms is important for the individualized therapy of irinotecan. Irinotecan 127-137 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 35-40 17558305-3 2007 Associations between UGT1A haplotypes and pharmacokinetics/pharmacodynamics of irinotecan were investigated to identify pharmacogenetic markers. Irinotecan 79-89 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 21-26 17558305-4 2007 METHODS: Associations between UGT1A haplotypes and the area under concentration curve ratio (SN-38 glucuronide/SN-38) or toxicities were analyzed in 177 Japanese cancer patients treated with irinotecan as a single agent or in combination chemotherapy. Irinotecan 93-98 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 30-35 17558305-10 2007 CONCLUSIONS: The haplotypes significantly associated with reduced area under concentration curve ratios and neutropenia contained UGT1A1*6 or *28, and both of them should be genotyped before irinotecan is given to Japanese and probably other Asian patients. Irinotecan 191-201 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 130-136 17609585-0 2007 Irinotecan activates p53 with its active metabolite, resulting in human hepatocellular carcinoma apoptosis. Irinotecan 0-10 tumor protein p53 Homo sapiens 21-24 17609585-4 2007 SN-38 significantly increased the expression of p53 protein and its phosphorylation at Ser(15) in the nucleus and apoptosis-inducing proteins Bax, caspase-9, and caspase-3, while it significantly decreased the antiapoptosis protein Bcl-xL of Huh7 cells. Irinotecan 0-5 tumor protein p53 Homo sapiens 48-51 17609585-4 2007 SN-38 significantly increased the expression of p53 protein and its phosphorylation at Ser(15) in the nucleus and apoptosis-inducing proteins Bax, caspase-9, and caspase-3, while it significantly decreased the antiapoptosis protein Bcl-xL of Huh7 cells. Irinotecan 0-5 BCL2 associated X, apoptosis regulator Homo sapiens 142-145 17609585-4 2007 SN-38 significantly increased the expression of p53 protein and its phosphorylation at Ser(15) in the nucleus and apoptosis-inducing proteins Bax, caspase-9, and caspase-3, while it significantly decreased the antiapoptosis protein Bcl-xL of Huh7 cells. Irinotecan 0-5 caspase 9 Homo sapiens 147-156 17609585-4 2007 SN-38 significantly increased the expression of p53 protein and its phosphorylation at Ser(15) in the nucleus and apoptosis-inducing proteins Bax, caspase-9, and caspase-3, while it significantly decreased the antiapoptosis protein Bcl-xL of Huh7 cells. Irinotecan 0-5 caspase 3 Homo sapiens 162-171 17609585-4 2007 SN-38 significantly increased the expression of p53 protein and its phosphorylation at Ser(15) in the nucleus and apoptosis-inducing proteins Bax, caspase-9, and caspase-3, while it significantly decreased the antiapoptosis protein Bcl-xL of Huh7 cells. Irinotecan 0-5 BCL2 like 1 Homo sapiens 232-238 17609585-5 2007 SN-38-induced apoptosis was recovered after p53 antisense oligodeoxynucleotide (AS ODN) pretreatment, while Huh7 cells were precultured with p53 AS ODN, followed by the addition of SN-38 for 24 h. Furthermore, increases in p53 DNA-binding activity were observed in the nuclei of Huh7 cells after SN-38 treatment as shown by electrophoretic mobility shift analysis. Irinotecan 0-5 tumor protein p53 Homo sapiens 44-47 17609585-5 2007 SN-38-induced apoptosis was recovered after p53 antisense oligodeoxynucleotide (AS ODN) pretreatment, while Huh7 cells were precultured with p53 AS ODN, followed by the addition of SN-38 for 24 h. Furthermore, increases in p53 DNA-binding activity were observed in the nuclei of Huh7 cells after SN-38 treatment as shown by electrophoretic mobility shift analysis. Irinotecan 0-5 tumor protein p53 Homo sapiens 141-144 17609585-5 2007 SN-38-induced apoptosis was recovered after p53 antisense oligodeoxynucleotide (AS ODN) pretreatment, while Huh7 cells were precultured with p53 AS ODN, followed by the addition of SN-38 for 24 h. Furthermore, increases in p53 DNA-binding activity were observed in the nuclei of Huh7 cells after SN-38 treatment as shown by electrophoretic mobility shift analysis. Irinotecan 0-5 tumor protein p53 Homo sapiens 141-144 17549342-1 2007 Tumor sensitivity to anticancer drugs such as CPT-11 and platinum derivatives was investigated by assessing Topo-1 and Bax/ERCC-1 expression in patients with stage I/II breast, lung, and gastric cancer who were positive for ONCs, and tumor sensitivity was compared between CPT-11 and platinum derivatives. Irinotecan 46-52 BCL2 associated X, apoptosis regulator Homo sapiens 119-122 17549342-1 2007 Tumor sensitivity to anticancer drugs such as CPT-11 and platinum derivatives was investigated by assessing Topo-1 and Bax/ERCC-1 expression in patients with stage I/II breast, lung, and gastric cancer who were positive for ONCs, and tumor sensitivity was compared between CPT-11 and platinum derivatives. Irinotecan 46-52 ERCC excision repair 1, endonuclease non-catalytic subunit Homo sapiens 123-129 17725105-0 2007 Schedule-dependent cytotoxicity of 5-fluorouracil and irinotecan in p53 mutant human colon cancer. Irinotecan 54-64 tumor protein p53 Homo sapiens 68-71 17516995-10 2007 Inhibitory effects of irinotecan on enzyme acetylcholinesterase (AChE) were studied in erythrocytes. Irinotecan 22-32 acetylcholinesterase (Cartwright blood group) Homo sapiens 43-63 17516995-10 2007 Inhibitory effects of irinotecan on enzyme acetylcholinesterase (AChE) were studied in erythrocytes. Irinotecan 22-32 acetylcholinesterase (Cartwright blood group) Homo sapiens 65-69 17516995-12 2007 Irinotecan was found to be strong inhibitor of the acetylcholine hydrolysis and to cause a continuous decrease of catalytic activity of AChE. Irinotecan 0-10 acetylcholinesterase (Cartwright blood group) Homo sapiens 136-140 17441964-5 2007 We also confirmed that the phosphorylated form of DNA-PK was increased by treatment with both etoposide and SN-38. Irinotecan 108-113 protein kinase, DNA-activated, catalytic subunit Homo sapiens 50-56 17510208-0 2007 UGT1A1 polymorphism can predict hematologic toxicity in patients treated with irinotecan. Irinotecan 78-88 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 17487378-0 2007 The interactive transcript abundance index [c-myc*p73alpha]/[p21*Bcl-2] correlates with baseline level of apoptosis and response to CPT-11 in human bronchogenic carcinoma cell lines. Irinotecan 132-138 MYC proto-oncogene, bHLH transcription factor Homo sapiens 44-49 17487378-0 2007 The interactive transcript abundance index [c-myc*p73alpha]/[p21*Bcl-2] correlates with baseline level of apoptosis and response to CPT-11 in human bronchogenic carcinoma cell lines. Irinotecan 132-138 BCL2 apoptosis regulator Homo sapiens 65-70 17487378-6 2007 In addition, the interactive transcript abundance index (ITAI) [c-myc*p73alpha]/[p21*Bcl-2] was directly correlated with baseline apoptosis (p<0.01) and CPT-11 response (p<0.05). Irinotecan 156-162 MYC proto-oncogene, bHLH transcription factor Homo sapiens 64-69 17487378-6 2007 In addition, the interactive transcript abundance index (ITAI) [c-myc*p73alpha]/[p21*Bcl-2] was directly correlated with baseline apoptosis (p<0.01) and CPT-11 response (p<0.05). Irinotecan 156-162 BCL2 apoptosis regulator Homo sapiens 85-90 17487378-7 2007 The ITAI was also correlated with CPT-11 response among cell lines derived from a variety of tissues that had inactivating p53 mutations or deletions, supporting its applicability for predicting response to camptothecins in other tissues regardless of p53 status. Irinotecan 34-40 tumor protein p53 Homo sapiens 123-126 17608024-0 2007 UGT1AI*6 and UGT1A1*27 for individualized irinotecan chemotherapy. Irinotecan 42-52 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 0-6 17608024-0 2007 UGT1AI*6 and UGT1A1*27 for individualized irinotecan chemotherapy. Irinotecan 42-52 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 13-19 17608024-1 2007 Genetic polymorphisms of uridine 5"-diphospho-glucuronosyl-transferase (UGT)1A1, a crucial drug-metabolizing enzyme of the anticancer drug irinotecan, are essential determinants of individual variation in susceptibility to irinotecan-related toxicity. Irinotecan 139-149 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 72-79 17608024-1 2007 Genetic polymorphisms of uridine 5"-diphospho-glucuronosyl-transferase (UGT)1A1, a crucial drug-metabolizing enzyme of the anticancer drug irinotecan, are essential determinants of individual variation in susceptibility to irinotecan-related toxicity. Irinotecan 223-233 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 72-79 17608024-4 2007 Therefore, it is reasonable to assume that the presence of UGT1A1*6 or UGTIA1*27 would also increase the risk of irinotecan toxicity. Irinotecan 113-123 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 59-65 17146732-12 2007 A DLT of grade 3 abdominal pain was seen at dose 600 mg BID of LY293111 with irinotecan 250 mg/m(2). Irinotecan 77-87 BH3 interacting domain death agonist Homo sapiens 56-59 17725105-2 2007 We evaluated cytotoxic effects of a sequentially administered a combination of 5-FU with CPT-11 in human p53 mutant colon cancer. Irinotecan 89-95 tumor protein p53 Homo sapiens 105-108 17575116-7 2007 Four minimally toxic compounds--BIM IV, BIM V, arcyriaflavin A, and K252c-reduced the relative resistance of ABCG2-transfected cells to SN-38 in cytotoxicity assays. Irinotecan 136-141 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 109-114 17534441-7 2007 In vivo, 4-IBP increased the antitumor effects of temozolomide and irinotecan in immunodeficient mice that were orthotopically grafted with invasive cancer cells. Irinotecan 67-77 translocator protein Mus musculus 11-14 17510416-0 2007 Copper-transporting P-type ATPase, ATP7A, confers multidrug resistance and its expression is related to resistance to SN-38 in clinical colon cancer. Irinotecan 118-123 ATPase copper transporting alpha Homo sapiens 35-40 17510416-5 2007 ATP7A expression also enhanced the efflux rates of doxorubicin and SN-38 from cells and increased the uptake of SN-38 in membrane vesicles. Irinotecan 67-72 ATPase copper transporting alpha Homo sapiens 0-5 17510416-5 2007 ATP7A expression also enhanced the efflux rates of doxorubicin and SN-38 from cells and increased the uptake of SN-38 in membrane vesicles. Irinotecan 112-117 ATPase copper transporting alpha Homo sapiens 0-5 17510416-8 2007 Using the histoculture drug response assay that is useful for the prediction of drug sensitivity of clinical cancers, ATP7A-expressing colon cancer cells were significantly more resistant to SN-38 than ATP7A-negative cells. Irinotecan 191-196 ATPase copper transporting alpha Homo sapiens 118-123 17513603-9 2007 Furthermore, c-FLIP gene silencing sensitized MDA435 cells to other chemotherapies, including etoposide, mitoxantrone, and SN-38. Irinotecan 123-128 CASP8 and FADD like apoptosis regulator Homo sapiens 13-19 17529881-4 2007 SN-38 is further metabolized to SN-38G by various hepatic and extrahepatic UGT1A isozymes, mainly UGT1A1. Irinotecan 0-5 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 75-80 17529881-4 2007 SN-38 is further metabolized to SN-38G by various hepatic and extrahepatic UGT1A isozymes, mainly UGT1A1. Irinotecan 0-5 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 98-104 17529881-4 2007 SN-38 is further metabolized to SN-38G by various hepatic and extrahepatic UGT1A isozymes, mainly UGT1A1. Irinotecan 32-37 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 75-80 17529881-4 2007 SN-38 is further metabolized to SN-38G by various hepatic and extrahepatic UGT1A isozymes, mainly UGT1A1. Irinotecan 32-37 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 98-104 17529881-5 2007 Impaired glucuronidation activity of the UGT1A1 enzyme has been linked with elevated levels of SN-38, leading to toxicities. Irinotecan 95-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 41-47 17529881-6 2007 UGT1A1*28 involves an extra TA repeat in the UGT1A1 promoter region and is the variant most frequently contributing to interpatient variability in irinotecan pharmacokinetics and toxicities. Irinotecan 147-157 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 17529881-6 2007 UGT1A1*28 involves an extra TA repeat in the UGT1A1 promoter region and is the variant most frequently contributing to interpatient variability in irinotecan pharmacokinetics and toxicities. Irinotecan 147-157 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 45-51 17529881-8 2007 Recently, UGT1A1*6 seems to contribute to the risk of toxicity of irinotecan in Asian patients. Irinotecan 66-76 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 10-16 17409690-8 2007 hCE2 preferentially hydrolyzes substrates with a small acyl moiety such as CPT-11, due to conformational steric hindrance in its active site. Irinotecan 75-81 carboxylesterase 2 Homo sapiens 0-4 17390033-2 2007 Gefitinib and imatinib are tyrosine kinase inhibitors which have clinical activities in several malignancies and they are also potent inhibitors of breast cancer resistance protein (BCRP) transporter, which confers the resistance of topoisomerase I inhibitors including SN-38 and topotecan. Irinotecan 270-275 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 148-180 17390033-2 2007 Gefitinib and imatinib are tyrosine kinase inhibitors which have clinical activities in several malignancies and they are also potent inhibitors of breast cancer resistance protein (BCRP) transporter, which confers the resistance of topoisomerase I inhibitors including SN-38 and topotecan. Irinotecan 270-275 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 182-186 17390033-10 2007 BCRP was equally overexpressed in the SBC-3/SN-38 with and without gefitinib or imatinib. Irinotecan 44-49 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 0-4 17390033-11 2007 In addition, the BCRP expression on the SBC-3/SN-38 cell membrane with and without gefitinib seemed to be equal. Irinotecan 46-51 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 17-21 17390039-1 2007 The sensitivity of LN metastases to anticancer drugs such as CPT-11 and platinum agents was investigated by assessing Topo-1 and Bax/ERCC-1 expression in patients who had stage III/Dukes" C colorectal cancer with ONCs. Irinotecan 61-67 BCL2 associated X, apoptosis regulator Homo sapiens 129-132 17390039-1 2007 The sensitivity of LN metastases to anticancer drugs such as CPT-11 and platinum agents was investigated by assessing Topo-1 and Bax/ERCC-1 expression in patients who had stage III/Dukes" C colorectal cancer with ONCs. Irinotecan 61-67 ERCC excision repair 1, endonuclease non-catalytic subunit Homo sapiens 133-139 17431115-7 2007 Thus, Apo2L/TRAIL + CPT-11 treatment-induced apoptosis is regulated through an S-phase checkpoint controlled by the Chk2-Cdc25A and Chk1-Cdc25A pathways and inhibition of Cdk2-associated kinase activity. Irinotecan 20-26 cyclin dependent kinase 2 Homo sapiens 171-175 17431115-8 2007 Low-dose CPT-11 and aphidicolin increased the proportion of S-phase cells and sensitized cells to Apo2L/TRAIL, by inducing phosphatidylserine externalization, caspase activation, and poly(ADP-ribose) polymerase 1 cleavage. Irinotecan 9-15 TNF superfamily member 10 Homo sapiens 98-103 17431115-8 2007 Low-dose CPT-11 and aphidicolin increased the proportion of S-phase cells and sensitized cells to Apo2L/TRAIL, by inducing phosphatidylserine externalization, caspase activation, and poly(ADP-ribose) polymerase 1 cleavage. Irinotecan 9-15 TNF superfamily member 10 Homo sapiens 104-109 17431115-8 2007 Low-dose CPT-11 and aphidicolin increased the proportion of S-phase cells and sensitized cells to Apo2L/TRAIL, by inducing phosphatidylserine externalization, caspase activation, and poly(ADP-ribose) polymerase 1 cleavage. Irinotecan 9-15 poly(ADP-ribose) polymerase 1 Homo sapiens 183-212 17391070-0 2007 Circulating VEGF reduction, response and outcome in advanced colorectal cancer patients treated with cetuximab plus irinotecan. Irinotecan 116-126 vascular endothelial growth factor A Homo sapiens 12-16 17391070-1 2007 OBJECTIVE: We designed this trial to investigate if modifications in levels of circulating vascular endothelial growth factor (VEGF) may be related to clinical response and outcome in advanced colorectal cancer patients during treatment with a weekly combination of cetuximab plus irinotecan. Irinotecan 281-291 vascular endothelial growth factor A Homo sapiens 91-125 17391070-1 2007 OBJECTIVE: We designed this trial to investigate if modifications in levels of circulating vascular endothelial growth factor (VEGF) may be related to clinical response and outcome in advanced colorectal cancer patients during treatment with a weekly combination of cetuximab plus irinotecan. Irinotecan 281-291 vascular endothelial growth factor A Homo sapiens 127-131 17391070-9 2007 CONCLUSION: These data represent the first evidence that suggests a role of VEGF reduction in the prediction of efficacy of treatment with cetuximab plus weekly irinotecan in heavily pretreated advanced colorectal cancer patients. Irinotecan 161-171 vascular endothelial growth factor A Homo sapiens 76-80 17395183-0 2007 Heat shock protein 27 is associated with irinotecan resistance in human colorectal cancer cells. Irinotecan 41-51 heat shock protein 90 beta family member 2, pseudogene Homo sapiens 0-18 17395183-2 2007 In the present study, we investigated the influence of Hsp expression on the irinotecan resistance of human colorectal cancer cells. Irinotecan 77-87 heat shock protein 90 beta family member 2, pseudogene Homo sapiens 55-58 17395183-3 2007 Among eight Hsp genes tested in this study, we confirmed that the expression of Hsp27 correlated with irinotecan resistance in colorectal cancer cells. Irinotecan 102-112 heat shock protein 90 beta family member 2, pseudogene Homo sapiens 12-15 17395183-3 2007 Among eight Hsp genes tested in this study, we confirmed that the expression of Hsp27 correlated with irinotecan resistance in colorectal cancer cells. Irinotecan 102-112 heat shock protein family B (small) member 1 Homo sapiens 80-85 17395183-4 2007 Specific inhibition of Hsp27 expression using an antisense oliogodeoxynucleotide increased the irinotecan sensitivity. Irinotecan 95-105 heat shock protein family B (small) member 1 Homo sapiens 23-28 17395183-5 2007 On the contrary, an overexpression of Hsp27 decreased the irinotecan sensitivity in colorectal cancer cells. Irinotecan 58-68 heat shock protein family B (small) member 1 Homo sapiens 38-43 17395183-7 2007 The expression level of Hsp27 determined by immunohistochemical analysis correlated with the clinical response to irinotecan in colorectal cancer patients. Irinotecan 114-124 heat shock protein family B (small) member 1 Homo sapiens 24-29 17395183-8 2007 Hsp27 expression levels of irinotecan-non-responder (mean staining score, 6.28; proportion of high staining score, 64.2%) were significantly higher compared to those of irinotecan-responder (mean staining score, 3.16; proportion of high staining score, 33.3%) (P for t-test=0.045). Irinotecan 27-37 heat shock protein family B (small) member 1 Homo sapiens 0-5 17395183-8 2007 Hsp27 expression levels of irinotecan-non-responder (mean staining score, 6.28; proportion of high staining score, 64.2%) were significantly higher compared to those of irinotecan-responder (mean staining score, 3.16; proportion of high staining score, 33.3%) (P for t-test=0.045). Irinotecan 169-179 heat shock protein family B (small) member 1 Homo sapiens 0-5 17395183-9 2007 These findings suggest that Hsp27 is involved in the irinotecan resistance of colorectal cancer cells possibly by reducing caspase-3 activity. Irinotecan 53-63 heat shock protein family B (small) member 1 Homo sapiens 28-33 17395183-9 2007 These findings suggest that Hsp27 is involved in the irinotecan resistance of colorectal cancer cells possibly by reducing caspase-3 activity. Irinotecan 53-63 caspase 3 Homo sapiens 123-132 17431115-2 2007 Previously, we reported that the cytotoxic effect of a zinc-bound form of Apo2 ligand/tumor necrosis factor-related apoptosis-inducing ligand (Apo2L/TRAIL), which is currently evaluated in clinical trials, in combination with low-dose CPT-11, induces apoptosis of C4-2 human prostate cancer cells and tissues. Irinotecan 235-241 TNF receptor superfamily member 10a Homo sapiens 74-78 17431115-2 2007 Previously, we reported that the cytotoxic effect of a zinc-bound form of Apo2 ligand/tumor necrosis factor-related apoptosis-inducing ligand (Apo2L/TRAIL), which is currently evaluated in clinical trials, in combination with low-dose CPT-11, induces apoptosis of C4-2 human prostate cancer cells and tissues. Irinotecan 235-241 TNF superfamily member 10 Homo sapiens 143-148 17431115-2 2007 Previously, we reported that the cytotoxic effect of a zinc-bound form of Apo2 ligand/tumor necrosis factor-related apoptosis-inducing ligand (Apo2L/TRAIL), which is currently evaluated in clinical trials, in combination with low-dose CPT-11, induces apoptosis of C4-2 human prostate cancer cells and tissues. Irinotecan 235-241 TNF superfamily member 10 Homo sapiens 149-154 17431115-7 2007 Thus, Apo2L/TRAIL + CPT-11 treatment-induced apoptosis is regulated through an S-phase checkpoint controlled by the Chk2-Cdc25A and Chk1-Cdc25A pathways and inhibition of Cdk2-associated kinase activity. Irinotecan 20-26 TNF superfamily member 10 Homo sapiens 6-11 17431115-7 2007 Thus, Apo2L/TRAIL + CPT-11 treatment-induced apoptosis is regulated through an S-phase checkpoint controlled by the Chk2-Cdc25A and Chk1-Cdc25A pathways and inhibition of Cdk2-associated kinase activity. Irinotecan 20-26 TNF superfamily member 10 Homo sapiens 12-17 17431115-7 2007 Thus, Apo2L/TRAIL + CPT-11 treatment-induced apoptosis is regulated through an S-phase checkpoint controlled by the Chk2-Cdc25A and Chk1-Cdc25A pathways and inhibition of Cdk2-associated kinase activity. Irinotecan 20-26 checkpoint kinase 2 Homo sapiens 116-120 17431115-7 2007 Thus, Apo2L/TRAIL + CPT-11 treatment-induced apoptosis is regulated through an S-phase checkpoint controlled by the Chk2-Cdc25A and Chk1-Cdc25A pathways and inhibition of Cdk2-associated kinase activity. Irinotecan 20-26 cell division cycle 25A Homo sapiens 121-127 17431115-7 2007 Thus, Apo2L/TRAIL + CPT-11 treatment-induced apoptosis is regulated through an S-phase checkpoint controlled by the Chk2-Cdc25A and Chk1-Cdc25A pathways and inhibition of Cdk2-associated kinase activity. Irinotecan 20-26 checkpoint kinase 1 Homo sapiens 132-136 17431115-7 2007 Thus, Apo2L/TRAIL + CPT-11 treatment-induced apoptosis is regulated through an S-phase checkpoint controlled by the Chk2-Cdc25A and Chk1-Cdc25A pathways and inhibition of Cdk2-associated kinase activity. Irinotecan 20-26 cell division cycle 25A Homo sapiens 137-143 17410034-3 2007 Genetic polymorphisms in the UGT1A1 gene have been identified as one of the causes of the increased diarrhea seen in white patients treated with irinotecan when compared with Japanese patients. Irinotecan 145-155 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 29-35 17531109-0 2007 Reversible grade 4 hyperbilirubinemia in a patient with UGT1A1 7/7 genotype treated with irinotecan and cetuximab. Irinotecan 89-99 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 56-62 17531109-5 2007 This case outlines the possibility of severe hepatic toxicity with moderate doses of irinotecan in patients with a UGT1A1 7/7 genotype. Irinotecan 85-95 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 115-121 17471158-6 2007 The anticancer agent irinotecan is metabolized to the active SN-38, which is further glucuronidated and detoxified by UGT 1A1. Irinotecan 21-31 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 118-125 17471158-6 2007 The anticancer agent irinotecan is metabolized to the active SN-38, which is further glucuronidated and detoxified by UGT 1A1. Irinotecan 61-66 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 118-125 17159606-0 2007 Role of p53 in irinotecan-induced intestinal cell death and mucosal damage. Irinotecan 15-25 Wistar clone pR53P1 p53 pseudogene Rattus norvegicus 8-11 17159606-3 2007 The aim of this study was to investigate the role of the p53 protein in the onset of intestinal damage following irinotecan treatment in two different settings. Irinotecan 113-123 Wistar clone pR53P1 p53 pseudogene Rattus norvegicus 57-60 17159606-10 2007 Irinotecan may act through upregulation of proapoptotic proteins Bax and Bak to induce cell death. Irinotecan 0-10 BCL2 associated X, apoptosis regulator Rattus norvegicus 65-68 16799812-12 2007 RT-PCR was used to confirm effects of irinotecan on caspase-1 expression in jejunal tissue and was significantly increased 6 h after treatment with irinotecan. Irinotecan 38-48 caspase 1 Rattus norvegicus 52-61 16799812-12 2007 RT-PCR was used to confirm effects of irinotecan on caspase-1 expression in jejunal tissue and was significantly increased 6 h after treatment with irinotecan. Irinotecan 148-158 caspase 1 Rattus norvegicus 52-61 17465213-0 2007 CPT-11 (SN-38) chemotherapy may be selectively applicable to biliary tract cancer with low hMLH1 expression. Irinotecan 0-6 mutL homolog 1 Homo sapiens 91-96 17465213-0 2007 CPT-11 (SN-38) chemotherapy may be selectively applicable to biliary tract cancer with low hMLH1 expression. Irinotecan 8-13 mutL homolog 1 Homo sapiens 91-96 17465213-8 2007 The hMLH1 expression was correlated with the IC50 for SN-38, although the relationship was not statistically significant (R = 0.717, p = 0.0715). Irinotecan 54-59 mutL homolog 1 Homo sapiens 4-9 17465213-11 2007 The cell growth of the hMLH1 dsRNA transfected group was decreased by approximately 50% by SN-38 exposure. Irinotecan 91-96 mutL homolog 1 Homo sapiens 23-28 17465213-14 2007 In conclusion, a low expression of hMLH1 in biliary tract cancer may aid in predicting its responsiveness to CPT-11 (SN38). Irinotecan 109-115 mutL homolog 1 Homo sapiens 35-40 17323127-5 2007 ABCG2 is the most recently described of the three major multidrug-resistance pumps, and its substrates include mitoxantrone, topotecan, irinotecan, flavopiridol, and methotrexate. Irinotecan 136-146 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 0-5 17367069-8 2007 In the animal model, irinotecan treatment led to a statistically significant decrease in tumor growth that was accompanied by a decrease in Bcl-2 and survivin levels and an increase in apoptotic cell death. Irinotecan 21-31 BCL2 apoptosis regulator Homo sapiens 140-145 17273745-1 2007 The aim of this study was to investigate the influence of combining thymidylate synthase (TS), X-ray cross complementing factor 1 (XRCC1) and uridine diphosphate glucoronosyltransferase (UGT1A1 *28) polymorphism genotypes in response rate and time to progression (TTP) in metastatic colorectal cancer patients treated with 5-fluorouracil (5-FU) plus irinotecan or oxaliplatin (OXA). Irinotecan 350-360 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 187-193 17159606-10 2007 Irinotecan may act through upregulation of proapoptotic proteins Bax and Bak to induce cell death. Irinotecan 0-10 BCL2-antagonist/killer 1 Rattus norvegicus 73-76 17255282-6 2007 CHIR-124 interacts synergistically with topoisomerase poisons (e.g., camptothecin or SN-38) in causing growth inhibition in several p53-mutant solid tumor cell lines as determined by isobologram or response surface analysis. Irinotecan 85-90 tumor protein p53 Homo sapiens 132-135 17965001-4 2007 The clinical application currently the most successful is the association of the anti-EGFR monoclonal antibody cetuximab with irinotecan in metastatic colorectal cancer. Irinotecan 126-136 epidermal growth factor receptor Homo sapiens 86-90 17713603-0 2007 Evaluation of HI-6 oxime: potential use in protection of human acetylcholinesterase inhibited by antineoplastic drug irinotecan and its cyto/genotoxicity in vitro. Irinotecan 117-127 acetylcholinesterase (Cartwright blood group) Homo sapiens 63-83 17713603-4 2007 Since their adverse effects are not well elucidated, in this study the efficiency of HI-6 oxime in protection and/or reactivation of human erythrocyte AChE inhibited by the antineoplastic drug irinotecan as well as its cyto/genotoxicity in vitro were investigated. Irinotecan 193-203 acetylcholinesterase (Cartwright blood group) Homo sapiens 151-155 17265738-3 2007 For example, the analysis of thiopurine S-methyltransferase genotypes enables the prediction of toxicity in patients to be treated with either 6-mercaptopurine or azathioprine, while the uridine 5"-diphosphoglucuronosyl-transferase 1A1 genotype may predict irinotecan toxicity. Irinotecan 257-267 thiopurine S-methyltransferase Homo sapiens 29-59 17185998-0 2007 Irinotecan-induced diarrhea: functional significance of the polymorphic ABCC2 transporter protein. Irinotecan 0-10 ATP binding cassette subfamily C member 2 Homo sapiens 72-77 17185998-2 2007 Life-threatening diarrhea is observed in up to 25% of patients receiving irinotecan and has been related with irinotecan pharmacokinetics and UGT1A1 genotype status. Irinotecan 73-83 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 142-148 17185998-3 2007 Here, we explore the association of ABCC2 (MRP2) polymorphisms and haplotypes with irinotecan disposition and diarrhea. Irinotecan 83-93 ATP binding cassette subfamily C member 2 Homo sapiens 36-41 17185998-6 2007 The haplotype ABCC2*2 was associated with lower irinotecan clearance (28.3 versus 31.6 l/h; P=0.020). Irinotecan 48-58 ATP binding cassette subfamily C member 2 Homo sapiens 14-19 17185998-9 2007 This study suggests that the presence of the ABCC2*2 haplotype is associated with less irinotecan-related diarrhea, maybe as a consequence of reduced hepatobiliary secretion of irinotecan. Irinotecan 87-97 ATP binding cassette subfamily C member 2 Homo sapiens 45-50 17185998-9 2007 This study suggests that the presence of the ABCC2*2 haplotype is associated with less irinotecan-related diarrhea, maybe as a consequence of reduced hepatobiliary secretion of irinotecan. Irinotecan 177-187 ATP binding cassette subfamily C member 2 Homo sapiens 45-50 17185998-10 2007 As the association was seen in patients not genetically predisposed at risk for diarrhea due to UGT1A1*28, confirmatory studies of the relationships of ABCC2 genotypes and irinotecan disposition and toxicity are warranted. Irinotecan 172-182 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 96-102 17185998-10 2007 As the association was seen in patients not genetically predisposed at risk for diarrhea due to UGT1A1*28, confirmatory studies of the relationships of ABCC2 genotypes and irinotecan disposition and toxicity are warranted. Irinotecan 172-182 ATP binding cassette subfamily C member 2 Homo sapiens 152-157 17520583-3 2007 Cetuximab, a chimeric mouse-human monoclonal antibody targeting the extracellular domain of the epidermal growth factor receptor (EGFR), has shown low but detectable activity when employed in pretreated patients either as a single agent or in combination with irinotecan. Irinotecan 260-270 epidermal growth factor receptor Homo sapiens 96-128 19356014-0 2007 ABCB1, SLCO1B1 and UGT1A1 gene polymorphisms are associated with toxicity in metastatic colorectal cancer patients treated with first-line irinotecan. Irinotecan 139-149 ATP binding cassette subfamily B member 1 Homo sapiens 0-5 19356014-0 2007 ABCB1, SLCO1B1 and UGT1A1 gene polymorphisms are associated with toxicity in metastatic colorectal cancer patients treated with first-line irinotecan. Irinotecan 139-149 solute carrier organic anion transporter family member 1B1 Homo sapiens 7-14 19356014-0 2007 ABCB1, SLCO1B1 and UGT1A1 gene polymorphisms are associated with toxicity in metastatic colorectal cancer patients treated with first-line irinotecan. Irinotecan 139-149 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 19-25 28207116-3 2007 Cetuximab, a chimeric mouse-human monoclonal antibody targeting the extracellular domain of the epidermal growth factor receptor (EGFR), has shown low but detectable activity when employed in pretreated patients either as a single agent or in combination with irinotecan. Irinotecan 260-270 epidermal growth factor receptor Homo sapiens 96-128 28207116-3 2007 Cetuximab, a chimeric mouse-human monoclonal antibody targeting the extracellular domain of the epidermal growth factor receptor (EGFR), has shown low but detectable activity when employed in pretreated patients either as a single agent or in combination with irinotecan. Irinotecan 260-270 epidermal growth factor receptor Homo sapiens 130-134 28207116-4 2007 Cetuximab in combination with irinotecan has been registered in the USA and Europe for the treatment of patients with metastatic colorectal cancer expressing the EGFR after failure of prior irinotecan-based cytotoxic therapy. Irinotecan 30-40 epidermal growth factor receptor Homo sapiens 162-166 28207119-3 2007 The chimeric anti- EGFR monoclonal antibody cetuximab has been approved for EGFR-expressing colorectal tumors in patients who progress after irinotecan-based chemotherapy in combination with irinotecan and in squamous cell head and neck carcinomas for patients with locally advanced disease in combination with radiation therapy or after failure of platinum-based chemotherapy in recurrent or metastatic disease (FDA). Irinotecan 141-151 epidermal growth factor receptor Homo sapiens 19-23 17520583-3 2007 Cetuximab, a chimeric mouse-human monoclonal antibody targeting the extracellular domain of the epidermal growth factor receptor (EGFR), has shown low but detectable activity when employed in pretreated patients either as a single agent or in combination with irinotecan. Irinotecan 260-270 epidermal growth factor receptor Homo sapiens 130-134 17520583-4 2007 Cetuximab in combination with irinotecan has been registered in the USA and Europe for the treatment of patients with metastatic colorectal cancer expressing the EGFR after failure of prior irinotecan-based cytotoxic therapy. Irinotecan 30-40 epidermal growth factor receptor Homo sapiens 162-166 17520586-3 2007 The chimeric anti- EGFR monoclonal antibody cetuximab has been approved for EGFR-expressing colorectal tumors in patients who progress after irinotecan-based chemotherapy in combination with irinotecan and in squamous cell head and neck carcinomas for patients with locally advanced disease in combination with radiation therapy or after failure of platinum-based chemotherapy in recurrent or metastatic disease (FDA). Irinotecan 141-151 epidermal growth factor receptor Homo sapiens 19-23 17913044-3 2007 In vivo delivery of irinotecan and floxuridine coencapsulated inside liposomes at the synergistic 1:1 molar ratio (referred to as CPX-1) lead to greatly enhanced efficacy compared to the two drugs administered as a saline-based cocktail in a number of human xenograft and murine tumor models. Irinotecan 20-30 complexin 1 Homo sapiens 130-135 17192505-3 2007 This analysis extended the work of a previous study that examined the effect of UGT1A1 genotypes on the incidence of severe neutropenia in 86 advanced cancer patients following irinotecan treatment. Irinotecan 177-187 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 80-86 17192505-7 2007 Based on these findings and other reports, the irinotecan label was modified to indicate the role of UGT1A1*28 polymorphism in the metabolism of irinotecan and the associated increased risk of severe neutropenia. Irinotecan 47-57 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 101-107 17192505-7 2007 Based on these findings and other reports, the irinotecan label was modified to indicate the role of UGT1A1*28 polymorphism in the metabolism of irinotecan and the associated increased risk of severe neutropenia. Irinotecan 145-155 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 101-107 17192505-8 2007 The label modifications also included recommendations for lower starting doses of irinotecan in patients homozygous for the UGT1A1*28 (7/7) polymorphism. Irinotecan 82-92 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 124-130 17552365-1 2007 We retrospectively analyzed the outcome of high-risk patients with non-small cell lung cancer (NSCLC) treated with nedaplatin (NP) and irinotecan (CPT) combination chemotherapy. Irinotecan 135-145 choline phosphotransferase 1 Homo sapiens 147-150 17913044-6 2007 Synergistic antitumor activity observed for CPX-1 was associated with maintenance of the 1:1 irinotecan:floxuridine molar ratio in plasma and tumor tissue over 16-24 h. In contrast, injection of the drugs combined in saline resulted in irinotecan:floxuridine ratios that changed 10-fold within 1 h in plasma and sevenfold within 4 h in tumor tissue. Irinotecan 93-103 complexin 1 Homo sapiens 44-49 17264798-1 2007 BACKGROUND: The conversion of CPT-11 to its active form, SN-38, by carboxylesterases (CESs) is a critical event in CPT-11-induced cytotoxicity. Irinotecan 30-36 carboxylesterase 1 Homo sapiens 67-84 17264798-2 2007 Among the CESs, CES1 and CES2 probably play a major role in the metabolism, but the functional significance and molecular basis of CES1 on CPT-11 response remain unclear. Irinotecan 139-145 carboxylesterase 1 Homo sapiens 131-135 17264798-1 2007 BACKGROUND: The conversion of CPT-11 to its active form, SN-38, by carboxylesterases (CESs) is a critical event in CPT-11-induced cytotoxicity. Irinotecan 57-62 carboxylesterase 1 Homo sapiens 67-84 17264798-1 2007 BACKGROUND: The conversion of CPT-11 to its active form, SN-38, by carboxylesterases (CESs) is a critical event in CPT-11-induced cytotoxicity. Irinotecan 115-121 carboxylesterase 1 Homo sapiens 67-84 17264798-3 2007 METHODS AND RESULTS: We investigated CES1A1 (AB119997) and CES1A2 (AB187225), whose coding sequences were recently registered in GenBank, for CPT-11-induced cytotoxicity, anticipating novel biomarkers of CPT-11 response. Irinotecan 142-148 carboxylesterase 1 pseudogene 1 Homo sapiens 59-65 17090741-5 2006 The labeling of irinotecan was recently changed and now includes a warning of greater neutropenia risk in patients with reduced activity in the drug-metabolizing enzyme uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1). Irinotecan 16-26 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 169-216 17106441-1 2006 This is the first phase II study of S-1 monotherapy for patients with metastatic colorectal cancer after failure of both irinotecan- and oxaliplatin-containing regimens. Irinotecan 121-131 proteasome 26S subunit, non-ATPase 1 Homo sapiens 36-39 17089053-2 2006 Our aim was to evaluate the effects of combination therapy with irinotecan hydrochloride and cisplatin (CPT-P), comparing to regimen with paclitaxel and platinum (TP). Irinotecan 64-88 ceramide-1-phosphate transfer protein Homo sapiens 104-109 17207039-2 2007 After receiving cisplatin/irinotecan, serum progastrin-releasing peptide (ProGRP) levels decreased to within the reference values and the lesions were markedly reduced in size. Irinotecan 26-36 gastrin releasing peptide Homo sapiens 44-72 17207039-2 2007 After receiving cisplatin/irinotecan, serum progastrin-releasing peptide (ProGRP) levels decreased to within the reference values and the lesions were markedly reduced in size. Irinotecan 26-36 gastrin releasing peptide Homo sapiens 74-80 17183650-5 2006 To test this hypothesis, we injected HB1.F3.C1 cells transduced to express an enzyme that efficiently activates the anti-cancer prodrug CPT-11 intravenously into mice bearing disseminated neuroblastoma tumors. Irinotecan 136-142 paternally expressed 10 Mus musculus 37-40 17183650-8 2006 Mice treated with the combination of HB1.F3.C1 cells expressing the CPT-11-activating enzyme and this prodrug produced tumor-free survival of 100% of the mice for >6 months (P<0.001 compared to control groups). Irinotecan 68-74 paternally expressed 10 Mus musculus 37-40 17148668-8 2006 Imatinib alone or in combination with irinotecan inhibited phosphorylation of PDGF-Rbeta of tumor-associated stromal cells and pericytes. Irinotecan 38-48 platelet derived growth factor receptor beta Homo sapiens 78-88 17184208-1 2006 Depending upon the UDP glucuronosyltransferase 1A1 (UGT1A1) genotype, patients are more or less susceptible to the risk of severe toxicity of irinotecan. Irinotecan 142-152 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 19-50 17184208-1 2006 Depending upon the UDP glucuronosyltransferase 1A1 (UGT1A1) genotype, patients are more or less susceptible to the risk of severe toxicity of irinotecan. Irinotecan 142-152 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 52-58 17184208-2 2006 As the US FDA-approved label of irinotecan (CPT-11, Camptosar) has been recently revised to include UGT1A1 genotype among potential risk factors for toxicity, it is expected that UGT1A1 genotyping will be increasingly used in patients undergoing irinotecan treatment. Irinotecan 32-42 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 179-185 17090741-5 2006 The labeling of irinotecan was recently changed and now includes a warning of greater neutropenia risk in patients with reduced activity in the drug-metabolizing enzyme uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1). Irinotecan 16-26 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 218-224 16894565-8 2006 High intratumoral mRNA levels of EGFR, ERCC1 and GSPT-P1 were each significantly associated with response to CPT-11 based chemotherapy. Irinotecan 109-115 epidermal growth factor receptor Homo sapiens 33-37 16894565-8 2006 High intratumoral mRNA levels of EGFR, ERCC1 and GSPT-P1 were each significantly associated with response to CPT-11 based chemotherapy. Irinotecan 109-115 ERCC excision repair 1, endonuclease non-catalytic subunit Homo sapiens 39-44 17090741-7 2006 Numerous studies have demonstrated the effects of genetic factors, especially UGT1A1*28, that contribute to interpatient variability in irinotecan pharmacokinetics and toxicity. Irinotecan 136-146 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 78-84 16894565-12 2006 This small retrospective study suggests that gene expression levels of EGFR, ERCC1 and GST-P1 may be useful in predicting the clinical outcome of patients with metastatic CRC treated with first-line CPT-11 based chemotherapy. Irinotecan 199-205 epidermal growth factor receptor Homo sapiens 71-75 17090741-8 2006 Irinotecan"s new labeling recommends that clinicians consider reducing the dosage of irinotecan in patients homozygous for UGT1A1*28. Irinotecan 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 123-129 16894565-12 2006 This small retrospective study suggests that gene expression levels of EGFR, ERCC1 and GST-P1 may be useful in predicting the clinical outcome of patients with metastatic CRC treated with first-line CPT-11 based chemotherapy. Irinotecan 199-205 ERCC excision repair 1, endonuclease non-catalytic subunit Homo sapiens 77-82 16894565-12 2006 This small retrospective study suggests that gene expression levels of EGFR, ERCC1 and GST-P1 may be useful in predicting the clinical outcome of patients with metastatic CRC treated with first-line CPT-11 based chemotherapy. Irinotecan 199-205 glutathione S-transferase pi 1 Homo sapiens 87-93 17090741-8 2006 Irinotecan"s new labeling recommends that clinicians consider reducing the dosage of irinotecan in patients homozygous for UGT1A1*28. Irinotecan 85-95 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 123-129 17090741-9 2006 CONCLUSION: At least part of the interpatient variability of irinotecan toxicity can be explained by the UGT1A1*28 polymorphism. Irinotecan 61-71 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 105-111 17090741-10 2006 Patients who are homozygous for the UGT1A1*28 allele have an increased risk of developing severe neutropenia when receiving irinotecan, especially the 300-350- mg/m2 regimen. Irinotecan 124-134 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 36-42 17185519-4 2006 The irinotecan/UGT1A1 issue and the development of molecular diagnostic testing are now to be translated into clinical practice. Irinotecan 4-14 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 15-21 16965601-4 2006 Pharmacogenetic relationships were explored between the UGT1A1 genotype and the ratio of the area under the plasma concentration-time curve (AUC) of the active metabolite of irinotecan (SN-38) to that of SN-38 glucuronide (SN-38G), used as a surrogate for UGT1A1 activity (AUC(SN-38)/AUC(SN-38G)). Irinotecan 204-209 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 56-62 17185519-1 2006 This article focuses on pharmacogenetic associations between genetic polymorphism of uridine diphosphate glucuronosyltransferase (UGT) 1A1 gene and irinotecan toxicity. Irinotecan 148-158 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 85-138 17185519-3 2006 On the basis of these backgrounds, the irinotecan label was updated in 2005 in the United States to provide pharmacogenetic information, and a dose reduction of irinotecan should be considered for patients known to be homozygous for the UGT1A1*28 allele when administered in combination with other agents or a single agent. Irinotecan 161-171 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 237-243 16965601-4 2006 Pharmacogenetic relationships were explored between the UGT1A1 genotype and the ratio of the area under the plasma concentration-time curve (AUC) of the active metabolite of irinotecan (SN-38) to that of SN-38 glucuronide (SN-38G), used as a surrogate for UGT1A1 activity (AUC(SN-38)/AUC(SN-38G)). Irinotecan 204-209 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 56-62 16965601-4 2006 Pharmacogenetic relationships were explored between the UGT1A1 genotype and the ratio of the area under the plasma concentration-time curve (AUC) of the active metabolite of irinotecan (SN-38) to that of SN-38 glucuronide (SN-38G), used as a surrogate for UGT1A1 activity (AUC(SN-38)/AUC(SN-38G)). Irinotecan 204-209 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 56-62 16965601-10 2006 The two patients simultaneously heterozygous for UGT1A1*28 and UGT1A1*6 and the two patients homozygous for UGT1A1*6 had significantly higher AUC(SN-38)/AUC(SN-38G) ratios than the others (P = 0.0039). Irinotecan 146-151 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 49-55 16965601-10 2006 The two patients simultaneously heterozygous for UGT1A1*28 and UGT1A1*6 and the two patients homozygous for UGT1A1*6 had significantly higher AUC(SN-38)/AUC(SN-38G) ratios than the others (P = 0.0039). Irinotecan 146-151 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 63-69 16965601-10 2006 The two patients simultaneously heterozygous for UGT1A1*28 and UGT1A1*6 and the two patients homozygous for UGT1A1*6 had significantly higher AUC(SN-38)/AUC(SN-38G) ratios than the others (P = 0.0039). Irinotecan 146-151 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 63-69 16965601-10 2006 The two patients simultaneously heterozygous for UGT1A1*28 and UGT1A1*6 and the two patients homozygous for UGT1A1*6 had significantly higher AUC(SN-38)/AUC(SN-38G) ratios than the others (P = 0.0039). Irinotecan 157-162 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 49-55 16965601-11 2006 Concurrence of UGT1A1*28 and UGT1A1*6, even when heterozygous, altered the disposition of irinotecan remarkably, potentially increasing susceptibility to toxicity. Irinotecan 90-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 15-21 16965601-11 2006 Concurrence of UGT1A1*28 and UGT1A1*6, even when heterozygous, altered the disposition of irinotecan remarkably, potentially increasing susceptibility to toxicity. Irinotecan 90-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 29-35 16965601-13 2006 UGT1A1 polymorphisms in the coding region of the UGT1A1 gene should be genotyped in addition to testing for UGT1A1*28 to more accurately predict irinotecan-related toxicity, at least in Asian patients. Irinotecan 145-155 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 16965601-13 2006 UGT1A1 polymorphisms in the coding region of the UGT1A1 gene should be genotyped in addition to testing for UGT1A1*28 to more accurately predict irinotecan-related toxicity, at least in Asian patients. Irinotecan 145-155 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 49-55 16965601-13 2006 UGT1A1 polymorphisms in the coding region of the UGT1A1 gene should be genotyped in addition to testing for UGT1A1*28 to more accurately predict irinotecan-related toxicity, at least in Asian patients. Irinotecan 145-155 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 49-55 16965601-0 2006 Pharmacogenetic impact of polymorphisms in the coding region of the UGT1A1 gene on SN-38 glucuronidation in Japanese patients with cancer. Irinotecan 83-88 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 68-74 16965601-1 2006 Pharmacogenetic testing for UDP-glucuronosyltransferase (UGT) 1A1*28, a promoter variant of the UGT1A1 gene, is now carried out clinically to estimate the risk of irinotecan-associated toxicity. Irinotecan 163-173 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 28-65 16965601-1 2006 Pharmacogenetic testing for UDP-glucuronosyltransferase (UGT) 1A1*28, a promoter variant of the UGT1A1 gene, is now carried out clinically to estimate the risk of irinotecan-associated toxicity. Irinotecan 163-173 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 96-102 16965601-4 2006 Pharmacogenetic relationships were explored between the UGT1A1 genotype and the ratio of the area under the plasma concentration-time curve (AUC) of the active metabolite of irinotecan (SN-38) to that of SN-38 glucuronide (SN-38G), used as a surrogate for UGT1A1 activity (AUC(SN-38)/AUC(SN-38G)). Irinotecan 174-184 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 56-62 16965601-4 2006 Pharmacogenetic relationships were explored between the UGT1A1 genotype and the ratio of the area under the plasma concentration-time curve (AUC) of the active metabolite of irinotecan (SN-38) to that of SN-38 glucuronide (SN-38G), used as a surrogate for UGT1A1 activity (AUC(SN-38)/AUC(SN-38G)). Irinotecan 186-191 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 56-62 29788595-0 2006 Novel personalized medicine technology: UGT1A1 testing for irinotecan as a case study. Irinotecan 59-69 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 40-46 17016579-3 2006 We present a colon cancer patient with the UGT1A1 polymorphism (UGT1A1 *28) as a known high risk for irinotecan, who was treated with a combination of doxifluridine and irinotecan for peritoneal dissemination resulting in stable disease for 2 years without adverse reactions, although the patient initially developed severe adverse effects to the combination of the protracted venous infusion of 5-FU and irinotecan. Irinotecan 101-111 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 43-49 17016579-3 2006 We present a colon cancer patient with the UGT1A1 polymorphism (UGT1A1 *28) as a known high risk for irinotecan, who was treated with a combination of doxifluridine and irinotecan for peritoneal dissemination resulting in stable disease for 2 years without adverse reactions, although the patient initially developed severe adverse effects to the combination of the protracted venous infusion of 5-FU and irinotecan. Irinotecan 101-111 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 64-70 17016579-3 2006 We present a colon cancer patient with the UGT1A1 polymorphism (UGT1A1 *28) as a known high risk for irinotecan, who was treated with a combination of doxifluridine and irinotecan for peritoneal dissemination resulting in stable disease for 2 years without adverse reactions, although the patient initially developed severe adverse effects to the combination of the protracted venous infusion of 5-FU and irinotecan. Irinotecan 169-179 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 43-49 17016579-3 2006 We present a colon cancer patient with the UGT1A1 polymorphism (UGT1A1 *28) as a known high risk for irinotecan, who was treated with a combination of doxifluridine and irinotecan for peritoneal dissemination resulting in stable disease for 2 years without adverse reactions, although the patient initially developed severe adverse effects to the combination of the protracted venous infusion of 5-FU and irinotecan. Irinotecan 169-179 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 64-70 17016579-3 2006 We present a colon cancer patient with the UGT1A1 polymorphism (UGT1A1 *28) as a known high risk for irinotecan, who was treated with a combination of doxifluridine and irinotecan for peritoneal dissemination resulting in stable disease for 2 years without adverse reactions, although the patient initially developed severe adverse effects to the combination of the protracted venous infusion of 5-FU and irinotecan. Irinotecan 169-179 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 43-49 17016579-3 2006 We present a colon cancer patient with the UGT1A1 polymorphism (UGT1A1 *28) as a known high risk for irinotecan, who was treated with a combination of doxifluridine and irinotecan for peritoneal dissemination resulting in stable disease for 2 years without adverse reactions, although the patient initially developed severe adverse effects to the combination of the protracted venous infusion of 5-FU and irinotecan. Irinotecan 169-179 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 64-70 29788595-1 2006 Third Wave"s Invader UDP glucuronosyltransferase 1A1 (UGT1A1) Molecular Assay, a genotyping system to predict adverse drug reactions in patients receiving the chemotherapeutic agent irinotecan (Camptosar , Pzifer, NY, USA) for the treatment of metastatic colorectal cancer (mCRC), was recently approved by the US FDA. Irinotecan 183-193 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 22-53 29788595-1 2006 Third Wave"s Invader UDP glucuronosyltransferase 1A1 (UGT1A1) Molecular Assay, a genotyping system to predict adverse drug reactions in patients receiving the chemotherapeutic agent irinotecan (Camptosar , Pzifer, NY, USA) for the treatment of metastatic colorectal cancer (mCRC), was recently approved by the US FDA. Irinotecan 183-193 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 55-61 29788595-1 2006 Third Wave"s Invader UDP glucuronosyltransferase 1A1 (UGT1A1) Molecular Assay, a genotyping system to predict adverse drug reactions in patients receiving the chemotherapeutic agent irinotecan (Camptosar , Pzifer, NY, USA) for the treatment of metastatic colorectal cancer (mCRC), was recently approved by the US FDA. Irinotecan 195-204 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 55-61 29788595-3 2006 This descriptive analysis highlight these data and the issues for the translation of this test to practice, including gaps in the evidence base, issues regarding adoption of the test to clinical practice and the potential societal impact of UGT1A1 testing for irinotecan prescribing. Irinotecan 260-270 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 241-247 16437251-2 2006 We have demonstrated from a rat model that intestinal beta-glucuronidase may play a key role in the development of CPT-11-induced delayed diarrhea by the deconjugation of the luminal SN-38 glucuronide, and the elimination of the intestinal microflora by antibiotics or dosing of TJ-14, a Kampo medicine that contains beta-glucuronidase inhibitor baicalin, exerted a protective effect. Irinotecan 115-121 glucuronidase, beta Rattus norvegicus 54-72 17047068-0 2006 Novel SN-38-incorporating polymeric micelles, NK012, eradicate vascular endothelial growth factor-secreting bulky tumors. Irinotecan 6-11 vascular endothelial growth factor A Homo sapiens 63-97 17047068-4 2006 The purpose of this study is to investigate the pharmacologic character of NK012 as an anticancer agent, especially in a vascular endothelial growth factor (VEGF)-secreting tumor model. Irinotecan 75-80 vascular endothelial growth factor A Homo sapiens 121-155 17047068-4 2006 The purpose of this study is to investigate the pharmacologic character of NK012 as an anticancer agent, especially in a vascular endothelial growth factor (VEGF)-secreting tumor model. Irinotecan 75-80 vascular endothelial growth factor A Homo sapiens 157-161 17047068-11 2006 In the drug distribution analysis, an increased accumulation of SN-38 in SBC-3/VEGF tumors was observed as compared with that in SBC-3/Neo tumors. Irinotecan 64-69 vascular endothelial growth factor A Homo sapiens 79-83 17047068-12 2006 NK012 markedly enhanced the antitumor activity of SN-38, especially in highly VEGF-secreting tumors, and could be a promising SN-38-based formulation. Irinotecan 50-55 vascular endothelial growth factor A Homo sapiens 78-82 16437251-2 2006 We have demonstrated from a rat model that intestinal beta-glucuronidase may play a key role in the development of CPT-11-induced delayed diarrhea by the deconjugation of the luminal SN-38 glucuronide, and the elimination of the intestinal microflora by antibiotics or dosing of TJ-14, a Kampo medicine that contains beta-glucuronidase inhibitor baicalin, exerted a protective effect. Irinotecan 115-121 glucuronidase, beta Rattus norvegicus 317-335 16896007-9 2006 Both 99mTc-DIDA/DISIDA and MIBI 1-hour RET correlated with SN-38 area under the curve (AUC; P < .01). Irinotecan 59-64 ret proto-oncogene Homo sapiens 39-42 16982469-3 2006 Recent FDA approval of genetic testing for mutations in the UGT1A1 gene that predict adverse reactions to irinotecan is ushering in a new era that will increasingly rely on genotyping to individualize treatment decisions for patients with cancer as well as for patients at high risk who may be candidates for chemoprevention agents. Irinotecan 106-116 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 60-66 17030643-2 2006 Monoclonal antibodies against the epidermal growth factor receptor (EGFR) are active against EGFR-expressing mCRC that is refractory to irinotecan. Irinotecan 136-146 epidermal growth factor receptor Homo sapiens 34-66 17030643-2 2006 Monoclonal antibodies against the epidermal growth factor receptor (EGFR) are active against EGFR-expressing mCRC that is refractory to irinotecan. Irinotecan 136-146 epidermal growth factor receptor Homo sapiens 68-72 17015060-0 2006 Effect of S-1 on pharmacokinetics of irinotecan in a patient with colorectal cancer. Irinotecan 37-47 proteasome 26S subunit, non-ATPase 1 Homo sapiens 10-13 16815871-0 2006 A mechanistic study on reduced toxicity of irinotecan by coadministered thalidomide, a tumor necrosis factor-alpha inhibitor. Irinotecan 43-53 tumor necrosis factor Rattus norvegicus 87-114 16815871-14 2006 Thalidomide and PGA also significantly inhibited P-glycoprotein (PgP/MDR1), multidrug resistance-associated protein (MRP1)- and MRP2-mediated CPT-11 and SN-38 transport in MDCKII cells. Irinotecan 142-148 ATP binding cassette subfamily C member 1 Canis lupus familiaris 76-121 16815871-14 2006 Thalidomide and PGA also significantly inhibited P-glycoprotein (PgP/MDR1), multidrug resistance-associated protein (MRP1)- and MRP2-mediated CPT-11 and SN-38 transport in MDCKII cells. Irinotecan 142-148 ATP binding cassette subfamily C member 2 Canis lupus familiaris 128-132 16815871-14 2006 Thalidomide and PGA also significantly inhibited P-glycoprotein (PgP/MDR1), multidrug resistance-associated protein (MRP1)- and MRP2-mediated CPT-11 and SN-38 transport in MDCKII cells. Irinotecan 153-158 ATP binding cassette subfamily C member 1 Canis lupus familiaris 76-121 16815871-14 2006 Thalidomide and PGA also significantly inhibited P-glycoprotein (PgP/MDR1), multidrug resistance-associated protein (MRP1)- and MRP2-mediated CPT-11 and SN-38 transport in MDCKII cells. Irinotecan 153-158 ATP binding cassette subfamily C member 2 Canis lupus familiaris 128-132 16896007-10 2006 On multiple regression analysis, SN-38 AUC = -215 + 18.68 x bilirubin + 4.27 x MIBI 1-hour RET (P = .009, R2 = 44.2%). Irinotecan 33-38 ret proto-oncogene Homo sapiens 91-94 16943524-0 2006 Thymidine phosphorylase expression is associated with response to capecitabine plus irinotecan in patients with metastatic colorectal cancer. Irinotecan 84-94 thymidine phosphorylase Homo sapiens 0-23 16404634-1 2006 PURPOSE: Several studies have demonstrated significant interactions between immunosuppressants (e.g., cyclosporin A) and chemotherapeutic drugs that are BCRP substrates (e.g., irinotecan), resulting in increased bioavailability and reduced clearance of these agents. Irinotecan 176-186 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 153-157 16969123-1 2006 BACKGROUND: Irinotecan is metabolized by various enzymes, including carboxylesterases, cytochrome P450 3A isozymes (CYP3A) and uridine-diphosphate glucuronosyltransferase 1A isoforms (UGT1A). Irinotecan 12-22 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 184-189 18666754-5 2006 Furthermore, the analysis of the conversion of irinotecan into SN-38 by carboxylesterase, the detoxification of irinotecan and SN-38 by CYP3A4 and UDP-glucuronosyl transferases, and the activity of excretory systems (i.e., cMOAT, P-gp and MRP) seems able to predict the interindividual variability in pharmacokinetics and pharmacodynamics, being possible to predict untolerable toxicities. Irinotecan 127-132 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 136-142 17086870-3 2006 The purpose of this study was to evaluate the cytotoxic effect of CPT-11 alone and in combination with mitomycin C (MMC) and hyperthermia on three colorectal cancer cell lines: CACO-2, HT-29, and DHD/K12/TRb (PROb). Irinotecan 66-72 T cell receptor beta locus Homo sapiens 204-207 16865249-8 2006 Our data suggest that GSTT1-null is associated with a greater probability of developing toxicity to 5-Fu/CPT-11/Lv treatments, indicating a potential application of this genetic analysis in predicting adverse effects of this regimen. Irinotecan 105-111 glutathione S-transferase theta 1 Homo sapiens 22-27 16951398-12 2006 It is suggested that the UGT1A1*28 genotype status could be used as a screening tool for a priori prevention of irinotecan-induced delayed-type diarrhea. Irinotecan 112-122 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 25-31 16842384-0 2006 The in vitro metabolism of irinotecan (CPT-11) by carboxylesterase and beta-glucuronidase in human colorectal tumours. Irinotecan 27-37 glucuronidase beta Homo sapiens 71-89 16545899-4 2006 7-Ethyl-10-hydroxycamptothecin (SN-38) glucuronidation, used as a probe for UGT1A1, showed sigmoidal kinetics with a Hill coefficient (n) of 1.2-1.3 in control and BNF-pretreated HepG2 cells. Irinotecan 0-30 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 76-82 16545899-4 2006 7-Ethyl-10-hydroxycamptothecin (SN-38) glucuronidation, used as a probe for UGT1A1, showed sigmoidal kinetics with a Hill coefficient (n) of 1.2-1.3 in control and BNF-pretreated HepG2 cells. Irinotecan 32-37 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 76-82 16906022-1 2006 OBJECTIVE: To investigate the pharmacogenetic effect of SLCO1B1 *1a, *1b, *5 and *15 polymorphisms on irinotecan disposition in Asian cancer patients. Irinotecan 102-112 solute carrier organic anion transporter family member 1B1 Homo sapiens 56-63 16906022-8 2006 CONCLUSION: These findings suggest that (1) SLCO1B1 haplotypes may have a significant influence on the disposition of irinotecan and its metabolites in Asian cancer patients, and (2) patients with SLCO1B1*15 haplotype may be susceptible to increased sensitivity to irinotecan, which may manifest itself either by increased efficacy or toxicity or both owing to the increased exposure levels to 7-ethyl-10-hydroxycamptothecin. Irinotecan 118-128 solute carrier organic anion transporter family member 1B1 Homo sapiens 44-51 16906022-8 2006 CONCLUSION: These findings suggest that (1) SLCO1B1 haplotypes may have a significant influence on the disposition of irinotecan and its metabolites in Asian cancer patients, and (2) patients with SLCO1B1*15 haplotype may be susceptible to increased sensitivity to irinotecan, which may manifest itself either by increased efficacy or toxicity or both owing to the increased exposure levels to 7-ethyl-10-hydroxycamptothecin. Irinotecan 118-128 solute carrier organic anion transporter family member 1B1 Homo sapiens 197-204 16906022-8 2006 CONCLUSION: These findings suggest that (1) SLCO1B1 haplotypes may have a significant influence on the disposition of irinotecan and its metabolites in Asian cancer patients, and (2) patients with SLCO1B1*15 haplotype may be susceptible to increased sensitivity to irinotecan, which may manifest itself either by increased efficacy or toxicity or both owing to the increased exposure levels to 7-ethyl-10-hydroxycamptothecin. Irinotecan 265-275 solute carrier organic anion transporter family member 1B1 Homo sapiens 44-51 16906022-8 2006 CONCLUSION: These findings suggest that (1) SLCO1B1 haplotypes may have a significant influence on the disposition of irinotecan and its metabolites in Asian cancer patients, and (2) patients with SLCO1B1*15 haplotype may be susceptible to increased sensitivity to irinotecan, which may manifest itself either by increased efficacy or toxicity or both owing to the increased exposure levels to 7-ethyl-10-hydroxycamptothecin. Irinotecan 265-275 solute carrier organic anion transporter family member 1B1 Homo sapiens 197-204 16906022-8 2006 CONCLUSION: These findings suggest that (1) SLCO1B1 haplotypes may have a significant influence on the disposition of irinotecan and its metabolites in Asian cancer patients, and (2) patients with SLCO1B1*15 haplotype may be susceptible to increased sensitivity to irinotecan, which may manifest itself either by increased efficacy or toxicity or both owing to the increased exposure levels to 7-ethyl-10-hydroxycamptothecin. Irinotecan 394-424 solute carrier organic anion transporter family member 1B1 Homo sapiens 44-51 16906022-8 2006 CONCLUSION: These findings suggest that (1) SLCO1B1 haplotypes may have a significant influence on the disposition of irinotecan and its metabolites in Asian cancer patients, and (2) patients with SLCO1B1*15 haplotype may be susceptible to increased sensitivity to irinotecan, which may manifest itself either by increased efficacy or toxicity or both owing to the increased exposure levels to 7-ethyl-10-hydroxycamptothecin. Irinotecan 394-424 solute carrier organic anion transporter family member 1B1 Homo sapiens 197-204 16809434-0 2006 Inhibition of poly(ADP-ribose) polymerase prevents irinotecan-induced intestinal damage and enhances irinotecan/temozolomide efficacy against colon carcinoma. Irinotecan 51-61 poly(ADP-ribose) polymerase 1 Homo sapiens 14-41 16809434-0 2006 Inhibition of poly(ADP-ribose) polymerase prevents irinotecan-induced intestinal damage and enhances irinotecan/temozolomide efficacy against colon carcinoma. Irinotecan 101-111 poly(ADP-ribose) polymerase 1 Homo sapiens 14-41 16809434-1 2006 Poly(ADP-ribose) polymerase (PARP) inhibitors enhance the antitumor activity of the topoisomerase I inhibitor irinotecan (CPT-11), which is used to treat advanced colorectal carcinoma. Irinotecan 110-120 poly(ADP-ribose) polymerase 1 Homo sapiens 0-27 16809434-1 2006 Poly(ADP-ribose) polymerase (PARP) inhibitors enhance the antitumor activity of the topoisomerase I inhibitor irinotecan (CPT-11), which is used to treat advanced colorectal carcinoma. Irinotecan 110-120 poly(ADP-ribose) polymerase 1 Homo sapiens 29-33 16809434-1 2006 Poly(ADP-ribose) polymerase (PARP) inhibitors enhance the antitumor activity of the topoisomerase I inhibitor irinotecan (CPT-11), which is used to treat advanced colorectal carcinoma. Irinotecan 122-128 poly(ADP-ribose) polymerase 1 Homo sapiens 0-27 16809434-1 2006 Poly(ADP-ribose) polymerase (PARP) inhibitors enhance the antitumor activity of the topoisomerase I inhibitor irinotecan (CPT-11), which is used to treat advanced colorectal carcinoma. Irinotecan 122-128 poly(ADP-ribose) polymerase 1 Homo sapiens 29-33 16809434-6 2006 Oral administration of GPI 15427 (40 mg/kg/q2x3 days) prevented intestinal injury and diarrhea induced by CPT-11 (30 mg/kg/day x 3 days) reducing inflammation and PARP-1 overactivation, as evidenced by immunohistochemical staining of intestinal tissue with antipoly(ADP-ribose) antibody (Ab). Irinotecan 106-112 poly(ADP-ribose) polymerase 1 Homo sapiens 163-169 16781005-3 2006 Therefore, we conducted a prospective feasibility study designed specifically to evaluate the efficacy of carboplatin (day 1) and irinotecan (days 1, 8, 15) with granulocyte colony-stimulating factor (G-CSF) support in elderly SCLC patients. Irinotecan 130-140 colony stimulating factor 3 Homo sapiens 162-199 16781005-3 2006 Therefore, we conducted a prospective feasibility study designed specifically to evaluate the efficacy of carboplatin (day 1) and irinotecan (days 1, 8, 15) with granulocyte colony-stimulating factor (G-CSF) support in elderly SCLC patients. Irinotecan 130-140 colony stimulating factor 3 Homo sapiens 201-206 16928820-5 2006 Nontoxic concentration of curcumin I, II, and III sensitized the ABCG2-expressing cells to mitoxantrone, topotecan, SN-38, and doxorubicin. Irinotecan 116-121 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 65-70 16857004-7 2006 After four courses of irinotecan, metastatic lesions were remarkably reduced in size, and the serum level of AFP decreased from 0.7 million to 927 ng/mL. Irinotecan 22-32 alpha fetoprotein Homo sapiens 109-112 16842384-0 2006 The in vitro metabolism of irinotecan (CPT-11) by carboxylesterase and beta-glucuronidase in human colorectal tumours. Irinotecan 39-45 glucuronidase beta Homo sapiens 71-89 16842384-4 2006 The aim of this study was to determine, the reactivation of SN-38 from SN-38 glucuronide by beta-glucuronidase may represent a significant pathway of SN-38 formation. Irinotecan 60-65 glucuronidase beta Homo sapiens 92-110 16842384-4 2006 The aim of this study was to determine, the reactivation of SN-38 from SN-38 glucuronide by beta-glucuronidase may represent a significant pathway of SN-38 formation. Irinotecan 71-76 glucuronidase beta Homo sapiens 92-110 16842384-6 2006 RESULTS: The rate of conversion of irinotecan (9.6 microm) was lower in tumour tissue than matched normal colon mucosa samples (0.30+/-0.14 pmol min-1 mg-1 protein and 0.77+/-0.59 pmol min-1 mg-1 protein, respectively; P<0.005). Irinotecan 35-45 CD59 molecule (CD59 blood group) Homo sapiens 145-155 16842384-6 2006 RESULTS: The rate of conversion of irinotecan (9.6 microm) was lower in tumour tissue than matched normal colon mucosa samples (0.30+/-0.14 pmol min-1 mg-1 protein and 0.77+/-0.59 pmol min-1 mg-1 protein, respectively; P<0.005). Irinotecan 35-45 CD59 molecule (CD59 blood group) Homo sapiens 185-195 16842384-9 2006 At equal concentrations of irinotecan and SN-38 glucuronide, the rate of beta-glucuronidase-mediated SN-38 production was higher than that formed from irinotecan in both tumour and normal tissue (P<0.05). Irinotecan 27-37 glucuronidase beta Homo sapiens 73-91 16842384-9 2006 At equal concentrations of irinotecan and SN-38 glucuronide, the rate of beta-glucuronidase-mediated SN-38 production was higher than that formed from irinotecan in both tumour and normal tissue (P<0.05). Irinotecan 42-47 glucuronidase beta Homo sapiens 73-91 16842384-9 2006 At equal concentrations of irinotecan and SN-38 glucuronide, the rate of beta-glucuronidase-mediated SN-38 production was higher than that formed from irinotecan in both tumour and normal tissue (P<0.05). Irinotecan 101-106 glucuronidase beta Homo sapiens 73-91 16842384-11 2006 CONCLUSIONS: Tumour beta-glucuronidase may play a significant role in the exposure of tumours to SN-38 in vivo. Irinotecan 97-102 glucuronidase beta Homo sapiens 20-38 16595709-0 2006 The novel UGT1A9 intronic I399 polymorphism appears as a predictor of 7-ethyl-10-hydroxycamptothecin glucuronidation levels in the liver. Irinotecan 70-100 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 10-16 16824027-0 2006 Screening for adverse reactions to irinotecan treatment using the Invader UGT1A1 Molecular Assay. Irinotecan 35-45 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 74-80 16824027-1 2006 Accumulating evidence provides support for the idea that determination of UGT1A1 polymorphisms before irinotecan (CPT-11) treatment is clinically useful and important for predicting and avoiding related toxicities. Irinotecan 102-112 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 74-80 16824027-1 2006 Accumulating evidence provides support for the idea that determination of UGT1A1 polymorphisms before irinotecan (CPT-11) treatment is clinically useful and important for predicting and avoiding related toxicities. Irinotecan 114-120 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 74-80 16824027-3 2006 A dose reduction of irinotecan should be considered for patients known to be homozygous for the UGT1A1*28 allele when administered in combination with other agents or as a single agent. Irinotecan 20-30 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 96-102 16824027-6 2006 This newly developed method for detecting UGT1A1 polymorphisms is feasible and has the potential to be widely used for rapid and accurate screening before irinotecan treatment. Irinotecan 155-165 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 42-48 16595709-4 2006 Of all the variants, the UGT1A9 I399C>T was associated with the most dramatic change in SN-38-glucuronide (SN-38G) (2.64-fold; p = 0.0007). Irinotecan 91-96 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 25-31 16595709-1 2006 Polymorphisms in UGT1A9 were associated with reduced toxicity and increased response to irinotecan in cancer patients. Irinotecan 88-98 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 17-23 16595709-6 2006 The very low linkage disequilibrium (r(2) < 0.19) between UGT1A9 I399 and all the other UGT1A1 and UGT1A9 variants suggests a direct effect or linkage to unknown functional variant(s) relevant to SN-38 glucuronidation. Irinotecan 199-204 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 61-67 16595709-7 2006 The UGT1A9 -118T(9/10) was also linked to alteration of SN-38 glucuronidation profiles in the liver (p < 0.05) and was associated with higher UGT1A1 protein (p = 0.03). Irinotecan 56-61 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 4-10 16713012-7 2006 When administered concurrently, gefitinib and SN-38 had a synergistic effect in five of the seven cell lines expressing wild-type EGFR, whereas the combination was antagonistic in PC-9 cells and a PC-9 subline resistant to gefitinib and expressing deletional mutant EGFR (PC-9/ZD). Irinotecan 46-51 epidermal growth factor receptor Homo sapiens 130-134 16595709-10 2006 Despite limitations resulting from sample size, results indicate that UGT1A9 I399 and -118T(9/10) may represent additional candidates in combination with UGT1A1 promoter haplotypes for the prediction of SN-38 glucuronidation profile in vivo. Irinotecan 203-208 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 70-76 16595709-10 2006 Despite limitations resulting from sample size, results indicate that UGT1A9 I399 and -118T(9/10) may represent additional candidates in combination with UGT1A1 promoter haplotypes for the prediction of SN-38 glucuronidation profile in vivo. Irinotecan 203-208 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 154-160 16809730-0 2006 The role of UGT1A1*28 polymorphism in the pharmacodynamics and pharmacokinetics of irinotecan in patients with metastatic colorectal cancer. Irinotecan 83-93 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-18 16809730-1 2006 PURPOSE: UGT1A1*28 polymorphism has been associated with decreased glucuronidation of SN38, the active metabolite of irinotecan. Irinotecan 117-127 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 9-15 16809730-11 2006 In particular, the possibility of a dose reduction for irinotecan in patients with a UGT1A1*28 polymorphism is not supported by the result of this analysis. Irinotecan 55-65 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 85-91 16713012-7 2006 When administered concurrently, gefitinib and SN-38 had a synergistic effect in five of the seven cell lines expressing wild-type EGFR, whereas the combination was antagonistic in PC-9 cells and a PC-9 subline resistant to gefitinib and expressing deletional mutant EGFR (PC-9/ZD). Irinotecan 46-51 epidermal growth factor receptor Homo sapiens 266-270 16713012-8 2006 When administered sequentially, treatment with SN-38 followed by gefitinib had remarkable synergistic effects in the PC-9 and PC-9/ZD cells. Irinotecan 47-52 proprotein convertase subtilisin/kexin type 9 Homo sapiens 117-121 16713012-8 2006 When administered sequentially, treatment with SN-38 followed by gefitinib had remarkable synergistic effects in the PC-9 and PC-9/ZD cells. Irinotecan 47-52 proprotein convertase subtilisin/kexin type 9 Homo sapiens 126-130 16723399-9 2006 Most importantly, WRN hypermethylation in colorectal tumors was a predictor of good clinical response to the camptothecin analogue irinotecan, a topoisomerase inhibitor commonly used in the clinical setting for the treatment of this tumor type. Irinotecan 131-141 WRN RecQ like helicase Homo sapiens 18-21 16897987-10 2006 It will be essential to prove that the combination of S-1 plus CPT-11 can replace the combination of infusional 5-FU and LV plus CPT-11 without negatively affecting efficacy and toxicity. Irinotecan 129-135 proteasome 26S subunit, non-ATPase 1 Homo sapiens 54-57 16897973-3 2006 S-1 based combination therapies with other promising drugs like cisplatin, irinotecan and taxanes, are expected to yield good results. Irinotecan 75-85 proteasome 26S subunit, non-ATPase 1 Homo sapiens 0-3 16897977-7 2006 Among various CPT-11 based chemotherapy, S-1 +CPT-11 appears to be the most effective and less toxic treatment. Irinotecan 14-20 proteasome 26S subunit, non-ATPase 1 Homo sapiens 41-52 16897989-3 2006 One of the combination therapies with S-1 plus irinotecan (CPT-11) has also been expected to have a better therapeutic value. Irinotecan 59-65 proteasome 26S subunit, non-ATPase 1 Homo sapiens 38-41 16187112-3 2006 The aim of this study was to ascertain whether two polymorphisms in the MTHFR gene (677C>T and 1298 A>C) could be used as genomic predictors of clinical response to fluoropyrimidine-based chemotherapy (in combination with irinotecan or oxaliplatin). Irinotecan 228-238 methylenetetrahydrofolate reductase Homo sapiens 72-77 16897968-5 2006 In combination with other anticancer drugs such as CPT-11 and taxanes, S-1 exercised synergistic antitumor efficacy not only on 5-FU-sensitive tumors with low expression levels of thymidylate synthase (TS) but also on 5-FU-resistant tumors with originally higher and/or elevated levels of TS expression. Irinotecan 51-57 proteasome 26S subunit, non-ATPase 1 Homo sapiens 71-74 16897968-6 2006 As one of the reasonable mechanism of antitumor synergism by the combination, CPT-11 and taxanes were found to reduce the expression of TS in human tumor resistant to 5-FU with high expression TS levels. Irinotecan 78-84 thymidylate synthetase Homo sapiens 136-138 16897968-6 2006 As one of the reasonable mechanism of antitumor synergism by the combination, CPT-11 and taxanes were found to reduce the expression of TS in human tumor resistant to 5-FU with high expression TS levels. Irinotecan 78-84 thymidylate synthetase Homo sapiens 193-195 16508919-5 2006 Many enzymes are involved in controlling the disposition of irinotecan, including the cellular target (TOP1), metabolism enzymes (CES2, UGT1A1, CYP3A4, CYP3A5), and cellular transporters of the anti-cancer agent (ABCB1, ABCC1, ABCC2, ABCC3, ABCC5, ABCG2). Irinotecan 60-70 carboxylesterase 2 Homo sapiens 130-134 16508919-5 2006 Many enzymes are involved in controlling the disposition of irinotecan, including the cellular target (TOP1), metabolism enzymes (CES2, UGT1A1, CYP3A4, CYP3A5), and cellular transporters of the anti-cancer agent (ABCB1, ABCC1, ABCC2, ABCC3, ABCC5, ABCG2). Irinotecan 60-70 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 136-142 16508919-5 2006 Many enzymes are involved in controlling the disposition of irinotecan, including the cellular target (TOP1), metabolism enzymes (CES2, UGT1A1, CYP3A4, CYP3A5), and cellular transporters of the anti-cancer agent (ABCB1, ABCC1, ABCC2, ABCC3, ABCC5, ABCG2). Irinotecan 60-70 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 144-150 16508919-5 2006 Many enzymes are involved in controlling the disposition of irinotecan, including the cellular target (TOP1), metabolism enzymes (CES2, UGT1A1, CYP3A4, CYP3A5), and cellular transporters of the anti-cancer agent (ABCB1, ABCC1, ABCC2, ABCC3, ABCC5, ABCG2). Irinotecan 60-70 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 152-158 16508919-5 2006 Many enzymes are involved in controlling the disposition of irinotecan, including the cellular target (TOP1), metabolism enzymes (CES2, UGT1A1, CYP3A4, CYP3A5), and cellular transporters of the anti-cancer agent (ABCB1, ABCC1, ABCC2, ABCC3, ABCC5, ABCG2). Irinotecan 60-70 ATP binding cassette subfamily B member 1 Homo sapiens 213-218 16508919-5 2006 Many enzymes are involved in controlling the disposition of irinotecan, including the cellular target (TOP1), metabolism enzymes (CES2, UGT1A1, CYP3A4, CYP3A5), and cellular transporters of the anti-cancer agent (ABCB1, ABCC1, ABCC2, ABCC3, ABCC5, ABCG2). Irinotecan 60-70 ATP binding cassette subfamily C member 1 Homo sapiens 220-225 16508919-5 2006 Many enzymes are involved in controlling the disposition of irinotecan, including the cellular target (TOP1), metabolism enzymes (CES2, UGT1A1, CYP3A4, CYP3A5), and cellular transporters of the anti-cancer agent (ABCB1, ABCC1, ABCC2, ABCC3, ABCC5, ABCG2). Irinotecan 60-70 ATP binding cassette subfamily C member 2 Homo sapiens 227-232 16508919-5 2006 Many enzymes are involved in controlling the disposition of irinotecan, including the cellular target (TOP1), metabolism enzymes (CES2, UGT1A1, CYP3A4, CYP3A5), and cellular transporters of the anti-cancer agent (ABCB1, ABCC1, ABCC2, ABCC3, ABCC5, ABCG2). Irinotecan 60-70 ATP binding cassette subfamily C member 3 Homo sapiens 234-239 16508919-5 2006 Many enzymes are involved in controlling the disposition of irinotecan, including the cellular target (TOP1), metabolism enzymes (CES2, UGT1A1, CYP3A4, CYP3A5), and cellular transporters of the anti-cancer agent (ABCB1, ABCC1, ABCC2, ABCC3, ABCC5, ABCG2). Irinotecan 60-70 ATP binding cassette subfamily C member 5 Homo sapiens 241-246 16508919-5 2006 Many enzymes are involved in controlling the disposition of irinotecan, including the cellular target (TOP1), metabolism enzymes (CES2, UGT1A1, CYP3A4, CYP3A5), and cellular transporters of the anti-cancer agent (ABCB1, ABCC1, ABCC2, ABCC3, ABCC5, ABCG2). Irinotecan 60-70 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 248-253 16794239-3 2006 DESIGN: This review summarizes current clinical data for EGFR-TKIs as monotherapy or in combination with 5-fluorouracil/leucovorin, irinotecan, or oxaliplatin, focusing on the rapidly developing area of colorectal, gastroesophageal, and pancreatic cancers. Irinotecan 132-142 epidermal growth factor receptor Homo sapiens 57-61 16818517-5 2006 We envisioned that dp-VP16 would be converted to the active chemotherapeutic agent VP-16 by the same rabbit carboxylesterase (rCE) that we have previously shown to efficiently activate the prodrug irinotecan (CPT-11). Irinotecan 197-207 host cell factor C1 Homo sapiens 22-26 16818517-5 2006 We envisioned that dp-VP16 would be converted to the active chemotherapeutic agent VP-16 by the same rabbit carboxylesterase (rCE) that we have previously shown to efficiently activate the prodrug irinotecan (CPT-11). Irinotecan 209-215 host cell factor C1 Homo sapiens 22-26 16818517-6 2006 This dp-VP16 prodrug might then be used in combination with CPT-11, with both drugs activated by a single enzyme. Irinotecan 60-66 host cell factor C1 Homo sapiens 8-12 16507564-3 2006 We designed this trial to investigate the modification of the vascular endothelial growth factor (VEGF) and interferon-gamma (IFN-gamma) in advanced colorectal cancer patients during treatment with a weekly combination of cetuximab plus irinotecan. Irinotecan 237-247 vascular endothelial growth factor A Homo sapiens 62-96 16501609-0 2006 Pleiotrophin, a candidate gene for poor tumor vasculature and in vivo neuroblastoma sensitivity to irinotecan. Irinotecan 99-109 pleiotrophin Homo sapiens 0-12 16501609-6 2006 PTN thus appeared to be a likely candidate gene associated with resistance to CPT-11 in this in vivo model. Irinotecan 78-84 pleiotrophin Homo sapiens 0-3 16501609-8 2006 PTN failed to demonstrate implication in resistance to CPT-11 in vitro but could influence sensitivity to CPT-11 exclusively through an in vivo mechanism. Irinotecan 106-112 pleiotrophin Homo sapiens 0-3 16636344-0 2006 Comprehensive analysis of UGT1A polymorphisms predictive for pharmacokinetics and treatment outcome in patients with non-small-cell lung cancer treated with irinotecan and cisplatin. Irinotecan 157-167 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 26-31 16636344-4 2006 We analyzed the association of UGT1A genotypes with irinotecan PK and clinical outcomes. Irinotecan 52-62 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 31-36 16636344-11 2006 CONCLUSION: These findings suggest that UGT1A1*6 and UGT1A9*22 genotypes may be important for SN-38 glucuronidation and associate with irinotecan-related severe toxicity. Irinotecan 94-99 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 40-46 16636344-11 2006 CONCLUSION: These findings suggest that UGT1A1*6 and UGT1A9*22 genotypes may be important for SN-38 glucuronidation and associate with irinotecan-related severe toxicity. Irinotecan 94-99 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 53-59 16636344-11 2006 CONCLUSION: These findings suggest that UGT1A1*6 and UGT1A9*22 genotypes may be important for SN-38 glucuronidation and associate with irinotecan-related severe toxicity. Irinotecan 135-145 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 40-46 16636344-11 2006 CONCLUSION: These findings suggest that UGT1A1*6 and UGT1A9*22 genotypes may be important for SN-38 glucuronidation and associate with irinotecan-related severe toxicity. Irinotecan 135-145 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 53-59 16636344-12 2006 Specifically, UGT1A1*6 might be useful for predicting tumor response and survival outcome of Korean patients with NSCLC treated with irinotecan-based chemotherapy. Irinotecan 133-143 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 14-20 16685247-0 2006 Invader UGT1A1 molecular assay for irinotecan toxicity. Irinotecan 35-45 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 8-14 16827119-6 2006 In contrast to the cells transfected with wild-type survivin, or the control NSCLC-3/neo, those cells transfected with mutant survivin and treated with SN-38 --> PS-341 exhibited enhanced caspase 9 activity (> 2-fold), caspase 3 (> 2- to 3-fold) activity and cytotoxicity compared to the NSCLC-3/neo cells. Irinotecan 152-157 caspase 9 Homo sapiens 191-200 16827119-6 2006 In contrast to the cells transfected with wild-type survivin, or the control NSCLC-3/neo, those cells transfected with mutant survivin and treated with SN-38 --> PS-341 exhibited enhanced caspase 9 activity (> 2-fold), caspase 3 (> 2- to 3-fold) activity and cytotoxicity compared to the NSCLC-3/neo cells. Irinotecan 152-157 caspase 3 Homo sapiens 225-234 16682733-4 2006 For example, pharmacogenetic tests currently used in cancer therapy, such as genotyping UGT1A1 to reduce the incidence of severe toxicity of irinotecan and sequencing epidermal growth factor receptor from tumors to identify somatic mutations conferring sensitivity to tyrosine kinase inhibitors, were developed without initial identification of interpopulation differences. Irinotecan 141-151 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 88-94 16641252-4 2006 An assay is available for genotypic testing of the enzyme UGT1A1, which is predictive of toxicity from irinotecan. Irinotecan 103-113 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 58-64 16821585-4 2006 RESULTS: Three independent lines of enquiry indicated that, in the tumour specimens, SN-38 was glucuronidated primarily by UGT1A1, the isozyme generally recognised as being responsible for hepatic detoxification of this compound, while with NU/ICRF 505 two candidate isoforms emerged - UGT1A8 and/or UGT1A10 - both of which are not normally expressed in the liver. Irinotecan 85-90 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 123-129 16821585-4 2006 RESULTS: Three independent lines of enquiry indicated that, in the tumour specimens, SN-38 was glucuronidated primarily by UGT1A1, the isozyme generally recognised as being responsible for hepatic detoxification of this compound, while with NU/ICRF 505 two candidate isoforms emerged - UGT1A8 and/or UGT1A10 - both of which are not normally expressed in the liver. Irinotecan 85-90 UDP glucuronosyltransferase family 1 member A8 Homo sapiens 286-292 16821585-4 2006 RESULTS: Three independent lines of enquiry indicated that, in the tumour specimens, SN-38 was glucuronidated primarily by UGT1A1, the isozyme generally recognised as being responsible for hepatic detoxification of this compound, while with NU/ICRF 505 two candidate isoforms emerged - UGT1A8 and/or UGT1A10 - both of which are not normally expressed in the liver. Irinotecan 85-90 UDP glucuronosyltransferase family 1 member A10 Homo sapiens 300-307 16507564-3 2006 We designed this trial to investigate the modification of the vascular endothelial growth factor (VEGF) and interferon-gamma (IFN-gamma) in advanced colorectal cancer patients during treatment with a weekly combination of cetuximab plus irinotecan. Irinotecan 237-247 interferon gamma Homo sapiens 108-124 16507564-3 2006 We designed this trial to investigate the modification of the vascular endothelial growth factor (VEGF) and interferon-gamma (IFN-gamma) in advanced colorectal cancer patients during treatment with a weekly combination of cetuximab plus irinotecan. Irinotecan 237-247 interferon gamma Homo sapiens 126-135 16494624-0 2006 Bcr-abl positive blast crisis of chronic myeloid leukemia emerging in a case of metastatic colorectal cancer 3 months after completion of an 8-month course of cetuximab and irinotecan. Irinotecan 173-183 ABL proto-oncogene 1, non-receptor tyrosine kinase Homo sapiens 0-7 16364925-4 2006 Interestingly, while the susceptibilities of Tbeta4 overexpressers to 5-FU and irinotecan remained unchanged, they were more resistant to doxorubicin and etoposide which triggered apoptosis via a mitochondrial pathway. Irinotecan 79-89 thymosin beta 4 X-linked Homo sapiens 45-51 16724931-2 2006 Recent clinical studies indicate that thalidomide, a known tumor necrosis factor-alpha inhibitor, ameliorated the toxicities induced by CPT-11. Irinotecan 136-142 tumor necrosis factor Rattus norvegicus 59-86 16446370-1 2006 DNA damage induced by the topoisomerase I inhibitor irinotecan (CPT-11) triggers in p53(WT) colorectal carcinoma cells a long term cell cycle arrest and in p53MUT cells a transient arrest followed by apoptosis (Magrini, R., Bhonde, M. R., Hanski, M. L., Notter, M., Scherubl, H., Boland, C. R., Zeitz, M., and Hanski, C. (2002) Int. Irinotecan 52-62 tumor protein p53 Homo sapiens 84-87 16618113-1 2006 ABCG2 is an ATP-binding cassette half-transporter conferring resistance to chemotherapeutic agents such as mitoxantrone, irinotecan, and flavopiridol. Irinotecan 121-131 ATP binding cassette subfamily B member 6 (Langereis blood group) Homo sapiens 12-49 16702730-1 2006 The ATP-binding cassette transporter, ABCG2, which is expressed at high levels in the intestine and liver, functions as an efflux transporter for many drugs, including clinically used anticancer agents such as topotecan and the active metabolite of irinotecan (SN-38). Irinotecan 249-259 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 38-43 16702730-1 2006 The ATP-binding cassette transporter, ABCG2, which is expressed at high levels in the intestine and liver, functions as an efflux transporter for many drugs, including clinically used anticancer agents such as topotecan and the active metabolite of irinotecan (SN-38). Irinotecan 261-266 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 38-43 16702730-2 2006 In this study, to elucidate the linkage disequilibrium (LD) profiles and haplotype structures of ABCG2, we have comprehensively searched for genetic variations in the putative promoter region, all the exons, and their flanking introns of ABCG2 from 177 Japanese cancer patients treated with irinotecan. Irinotecan 291-301 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 97-102 16525652-12 2006 in vivo results demonstrated that DPA enhanced the in vivo anticancer activity of the chemotherapeutic agent, CPT-11, by elevation of p53-independent p21/Cip1 and p27/Kip1 expression. Irinotecan 110-116 H3 histone pseudogene 16 Homo sapiens 150-153 16525652-12 2006 in vivo results demonstrated that DPA enhanced the in vivo anticancer activity of the chemotherapeutic agent, CPT-11, by elevation of p53-independent p21/Cip1 and p27/Kip1 expression. Irinotecan 110-116 cyclin dependent kinase inhibitor 1A Homo sapiens 154-158 16525652-12 2006 in vivo results demonstrated that DPA enhanced the in vivo anticancer activity of the chemotherapeutic agent, CPT-11, by elevation of p53-independent p21/Cip1 and p27/Kip1 expression. Irinotecan 110-116 interferon alpha inducible protein 27 Homo sapiens 163-166 16525652-12 2006 in vivo results demonstrated that DPA enhanced the in vivo anticancer activity of the chemotherapeutic agent, CPT-11, by elevation of p53-independent p21/Cip1 and p27/Kip1 expression. Irinotecan 110-116 cyclin dependent kinase inhibitor 1B Homo sapiens 167-171 16777630-6 2006 Moreover the association of the irinotecan with the anti EGFR antibody (Erbitux)), cetixumab) has show a efficacy in third line after progression under an protocol with irinotecan. Irinotecan 32-42 epidermal growth factor receptor Homo sapiens 57-61 16777630-6 2006 Moreover the association of the irinotecan with the anti EGFR antibody (Erbitux)), cetixumab) has show a efficacy in third line after progression under an protocol with irinotecan. Irinotecan 169-179 epidermal growth factor receptor Homo sapiens 57-61 16618113-1 2006 ABCG2 is an ATP-binding cassette half-transporter conferring resistance to chemotherapeutic agents such as mitoxantrone, irinotecan, and flavopiridol. Irinotecan 121-131 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 0-5 16518418-0 2006 Potentiation of irinotecan sensitivity by Se-methylselenocysteine in an in vivo tumor model is associated with downregulation of cyclooxygenase-2, inducible nitric oxide synthase, and hypoxia-inducible factor 1alpha expression, resulting in reduced angiogenesis. Irinotecan 16-26 prostaglandin-endoperoxide synthase 2 Homo sapiens 129-145 16518418-0 2006 Potentiation of irinotecan sensitivity by Se-methylselenocysteine in an in vivo tumor model is associated with downregulation of cyclooxygenase-2, inducible nitric oxide synthase, and hypoxia-inducible factor 1alpha expression, resulting in reduced angiogenesis. Irinotecan 16-26 nitric oxide synthase 2 Homo sapiens 147-178 16518418-0 2006 Potentiation of irinotecan sensitivity by Se-methylselenocysteine in an in vivo tumor model is associated with downregulation of cyclooxygenase-2, inducible nitric oxide synthase, and hypoxia-inducible factor 1alpha expression, resulting in reduced angiogenesis. Irinotecan 16-26 hypoxia inducible factor 1 subunit alpha Homo sapiens 184-215 16518418-3 2006 In FaDu xenografts, a poorly differentiated tumor-expressing mutant p53, the cure rate was increased from 30% with irinotecan alone to 100% with the combination of irinotecan and MSC. Irinotecan 115-125 tumor protein p53 Homo sapiens 68-71 16518418-3 2006 In FaDu xenografts, a poorly differentiated tumor-expressing mutant p53, the cure rate was increased from 30% with irinotecan alone to 100% with the combination of irinotecan and MSC. Irinotecan 164-174 tumor protein p53 Homo sapiens 68-71 16518418-7 2006 In contrast, significant downregulation of cyclooxygenase-2 (COX-2) expression and activity was observed in the cells exposed to SN-38 in combination with MSC compared to SN-38 alone. Irinotecan 129-134 prostaglandin-endoperoxide synthase 2 Homo sapiens 43-59 16518418-7 2006 In contrast, significant downregulation of cyclooxygenase-2 (COX-2) expression and activity was observed in the cells exposed to SN-38 in combination with MSC compared to SN-38 alone. Irinotecan 129-134 prostaglandin-endoperoxide synthase 2 Homo sapiens 61-66 16518418-7 2006 In contrast, significant downregulation of cyclooxygenase-2 (COX-2) expression and activity was observed in the cells exposed to SN-38 in combination with MSC compared to SN-38 alone. Irinotecan 171-176 prostaglandin-endoperoxide synthase 2 Homo sapiens 43-59 16518418-7 2006 In contrast, significant downregulation of cyclooxygenase-2 (COX-2) expression and activity was observed in the cells exposed to SN-38 in combination with MSC compared to SN-38 alone. Irinotecan 171-176 prostaglandin-endoperoxide synthase 2 Homo sapiens 61-66 16518418-9 2006 Analysis of tumor tissues at 24 h after treatment with synergistic modality of irinotecan and MSC revealed significant downregulation of COX-2, inducible nitric oxide synthase (iNOS) and hypoxia-induced factor-1alpha expression (HIF 1alpha). Irinotecan 79-89 prostaglandin-endoperoxide synthase 2 Homo sapiens 137-142 16518418-9 2006 Analysis of tumor tissues at 24 h after treatment with synergistic modality of irinotecan and MSC revealed significant downregulation of COX-2, inducible nitric oxide synthase (iNOS) and hypoxia-induced factor-1alpha expression (HIF 1alpha). Irinotecan 79-89 nitric oxide synthase 2 Homo sapiens 144-175 16518418-9 2006 Analysis of tumor tissues at 24 h after treatment with synergistic modality of irinotecan and MSC revealed significant downregulation of COX-2, inducible nitric oxide synthase (iNOS) and hypoxia-induced factor-1alpha expression (HIF 1alpha). Irinotecan 79-89 nitric oxide synthase 2 Homo sapiens 177-181 16518418-9 2006 Analysis of tumor tissues at 24 h after treatment with synergistic modality of irinotecan and MSC revealed significant downregulation of COX-2, inducible nitric oxide synthase (iNOS) and hypoxia-induced factor-1alpha expression (HIF 1alpha). Irinotecan 79-89 hypoxia inducible factor 1 subunit alpha Homo sapiens 229-239 16446370-1 2006 DNA damage induced by the topoisomerase I inhibitor irinotecan (CPT-11) triggers in p53(WT) colorectal carcinoma cells a long term cell cycle arrest and in p53MUT cells a transient arrest followed by apoptosis (Magrini, R., Bhonde, M. R., Hanski, M. L., Notter, M., Scherubl, H., Boland, C. R., Zeitz, M., and Hanski, C. (2002) Int. Irinotecan 52-62 tumor protein p53 Homo sapiens 156-159 16446370-1 2006 DNA damage induced by the topoisomerase I inhibitor irinotecan (CPT-11) triggers in p53(WT) colorectal carcinoma cells a long term cell cycle arrest and in p53MUT cells a transient arrest followed by apoptosis (Magrini, R., Bhonde, M. R., Hanski, M. L., Notter, M., Scherubl, H., Boland, C. R., Zeitz, M., and Hanski, C. (2002) Int. Irinotecan 64-70 tumor protein p53 Homo sapiens 84-87 16446370-1 2006 DNA damage induced by the topoisomerase I inhibitor irinotecan (CPT-11) triggers in p53(WT) colorectal carcinoma cells a long term cell cycle arrest and in p53MUT cells a transient arrest followed by apoptosis (Magrini, R., Bhonde, M. R., Hanski, M. L., Notter, M., Scherubl, H., Boland, C. R., Zeitz, M., and Hanski, C. (2002) Int. Irinotecan 64-70 tumor protein p53 Homo sapiens 156-159 16446370-5 2006 Here we used five p53WT and five p53MUT established colon carcinoma cell lines to identify gene expression alterations associated with apoptosis in p53MUT cells after treatment with SN-38, the irinotecan metabolite. Irinotecan 182-187 tumor protein p53 Homo sapiens 18-21 16446370-5 2006 Here we used five p53WT and five p53MUT established colon carcinoma cell lines to identify gene expression alterations associated with apoptosis in p53MUT cells after treatment with SN-38, the irinotecan metabolite. Irinotecan 182-187 tumor protein p53 Homo sapiens 33-36 16446370-10 2006 Together, these data indicate that the high expression of mitotic genes in p53MUT cells after SN-38 treatment contributes to DNA damage-induced apoptosis, whereas their suppression in p53WT cells acts as a safeguard mechanism preventing mitosis initiation and the subsequent apoptosis. Irinotecan 94-99 tumor protein p53 Homo sapiens 75-78 16309825-2 2006 However, overexpression of ABCG2 (BCRP/MXR/ABCP) reportedly confers cancer cells resistance to SN-38, the active form of CPT-11. Irinotecan 121-127 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 39-42 16551870-0 2006 Irinotecan inactivation is modulated by epigenetic silencing of UGT1A1 in colon cancer. Irinotecan 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 64-70 16551870-2 2006 The UGT1A1-metabolizing enzyme, expressed in liver and colon, is primarily involved in the inactivation of its active metabolite 7-ethyl-10-hydroxycamptothecin (SN-38). Irinotecan 129-159 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 16551870-2 2006 The UGT1A1-metabolizing enzyme, expressed in liver and colon, is primarily involved in the inactivation of its active metabolite 7-ethyl-10-hydroxycamptothecin (SN-38). Irinotecan 161-166 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 16551870-9 2006 Loss of UGT1A1 methylation was further associated with an increase in UGT1A1 protein content and with an enhanced inactivation of SN-38 by 300% in HCT-116 cells. Irinotecan 130-135 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 8-14 16551870-10 2006 CONCLUSIONS: We conclude that DNA methylation represses UGT1A1 expression in colon cancer and that this process may contribute to the level of tumoral inactivation of the anticancer agent SN-38 and potentially influence clinical response. Irinotecan 188-193 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 56-62 16303243-1 2006 Breast cancer resistance protein (BCRP) is a half-molecule ATP-binding cassette transporter that pumps out various anticancer agents such as 7-ethyl-10-hydroxycamptothecin, topotecan and mitoxantrone. Irinotecan 141-171 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 0-32 16303243-1 2006 Breast cancer resistance protein (BCRP) is a half-molecule ATP-binding cassette transporter that pumps out various anticancer agents such as 7-ethyl-10-hydroxycamptothecin, topotecan and mitoxantrone. Irinotecan 141-171 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 34-38 16309825-2 2006 However, overexpression of ABCG2 (BCRP/MXR/ABCP) reportedly confers cancer cells resistance to SN-38, the active form of CPT-11. Irinotecan 95-100 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 27-32 16309825-2 2006 However, overexpression of ABCG2 (BCRP/MXR/ABCP) reportedly confers cancer cells resistance to SN-38, the active form of CPT-11. Irinotecan 95-100 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 34-38 16309825-2 2006 However, overexpression of ABCG2 (BCRP/MXR/ABCP) reportedly confers cancer cells resistance to SN-38, the active form of CPT-11. Irinotecan 95-100 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 39-42 16309825-2 2006 However, overexpression of ABCG2 (BCRP/MXR/ABCP) reportedly confers cancer cells resistance to SN-38, the active form of CPT-11. Irinotecan 95-100 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 43-47 16309825-2 2006 However, overexpression of ABCG2 (BCRP/MXR/ABCP) reportedly confers cancer cells resistance to SN-38, the active form of CPT-11. Irinotecan 121-127 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 27-32 16309825-2 2006 However, overexpression of ABCG2 (BCRP/MXR/ABCP) reportedly confers cancer cells resistance to SN-38, the active form of CPT-11. Irinotecan 121-127 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 34-38 16309825-2 2006 However, overexpression of ABCG2 (BCRP/MXR/ABCP) reportedly confers cancer cells resistance to SN-38, the active form of CPT-11. Irinotecan 121-127 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 43-47 16343744-5 2006 In this review, the role of genetic polymorphisms in the main enzyme-systems (carboxylesterase, cytochrome P450 3A, and uridine diphosphate-glucuronosyltransferase) and ABC-transporters (ABCB1, ABCC2, and ABCG2) involved in irinotecan metabolism, are discussed. Irinotecan 224-234 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 120-163 16343744-5 2006 In this review, the role of genetic polymorphisms in the main enzyme-systems (carboxylesterase, cytochrome P450 3A, and uridine diphosphate-glucuronosyltransferase) and ABC-transporters (ABCB1, ABCC2, and ABCG2) involved in irinotecan metabolism, are discussed. Irinotecan 224-234 ATP binding cassette subfamily B member 1 Homo sapiens 187-192 16343744-5 2006 In this review, the role of genetic polymorphisms in the main enzyme-systems (carboxylesterase, cytochrome P450 3A, and uridine diphosphate-glucuronosyltransferase) and ABC-transporters (ABCB1, ABCC2, and ABCG2) involved in irinotecan metabolism, are discussed. Irinotecan 224-234 ATP binding cassette subfamily C member 2 Homo sapiens 194-199 16343744-5 2006 In this review, the role of genetic polymorphisms in the main enzyme-systems (carboxylesterase, cytochrome P450 3A, and uridine diphosphate-glucuronosyltransferase) and ABC-transporters (ABCB1, ABCC2, and ABCG2) involved in irinotecan metabolism, are discussed. Irinotecan 224-234 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 205-210 16619577-0 2006 Feasibility study of biweekly CPT-11 plus CDDP for S-1- and paclitaxel-refractory, metastatic gastric cancer. Irinotecan 30-36 proteasome 26S subunit, non-ATPase 1 Homo sapiens 51-54 16303861-0 2006 Cetuximab and irinotecan/5-fluorouracil/folinic acid is a safe combination for the first-line treatment of patients with epidermal growth factor receptor expressing metastatic colorectal carcinoma. Irinotecan 14-24 epidermal growth factor receptor Homo sapiens 121-153 16619577-9 2006 CONCLUSION: Biweekly CPT-11 plus CDDP was feasible for S-1- and paclitaxel-refractory metastatic gastric cancer, with moderate activity and favorable toxicity. Irinotecan 21-27 proteasome 26S subunit, non-ATPase 1 Homo sapiens 55-58 16542215-0 2006 Role of ABCG2 as a biomarker for predicting resistance to CPT-11/SN-38 in lung cancer. Irinotecan 58-64 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 8-13 16456808-1 2006 BACKGROUND: In the current Phase II study, the authors evaluated the association between genomic polymorphic variants in uridine diphosphate glucuronosyl transferase (UGT1A1), methylenetetrahydrofolate reductase (MTHFR), and thymidylate synthase (TS) genes, and the incidence of the adverse effects of irinotecan and raltitrexed in previously heavily treated patients with metastatic colorectal carcinoma. Irinotecan 302-312 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 167-173 16456808-10 2006 CONCLUSIONS: In the current study, UGT1A1 promoter polymorphism was found to be predictive of the risk of diarrhea, emesis, and fatigue caused by chemotherapy with irinotecan and raltitrexed. Irinotecan 164-174 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 35-41 16456808-11 2006 Screening for UGT1A1 promoter polymorphism may be clinically useful for identifying patients at a higher risk of developing a severe or potentially life-threatening toxicity after irinotecan-based chemotherapy. Irinotecan 180-190 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 14-20 16542215-7 2006 These results indicate that the upregulation of ABCG2 was functional, and related to the resistance of H23/SN-38 cells to SN-38. Irinotecan 107-112 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 48-53 16542215-0 2006 Role of ABCG2 as a biomarker for predicting resistance to CPT-11/SN-38 in lung cancer. Irinotecan 65-70 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 8-13 16542215-7 2006 These results indicate that the upregulation of ABCG2 was functional, and related to the resistance of H23/SN-38 cells to SN-38. Irinotecan 122-127 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 48-53 16541742-6 2006 The standard chemotherapy for extensive SCLC is the combination of cisplatin and irinotecan or etoposide. Irinotecan 81-91 SCLC1 Homo sapiens 40-44 16542215-8 2006 Moreover, we found that gene expression levels of ABCG2 were significantly correlated with the concentration of SN-38 for 50% cell survival in 13 NSCLC cells (r=0.592, P<0.05). Irinotecan 112-117 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 50-55 16542215-9 2006 The present results indicate that the induction of ABCG2 by SN-38 does confer acquired resistance to CPT-11/SN-38, but the induction of ABCG2 and subsequent drug resistance are reversible. Irinotecan 60-65 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 51-56 16542215-9 2006 The present results indicate that the induction of ABCG2 by SN-38 does confer acquired resistance to CPT-11/SN-38, but the induction of ABCG2 and subsequent drug resistance are reversible. Irinotecan 101-107 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 51-56 16542215-9 2006 The present results indicate that the induction of ABCG2 by SN-38 does confer acquired resistance to CPT-11/SN-38, but the induction of ABCG2 and subsequent drug resistance are reversible. Irinotecan 108-113 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 51-56 16542215-10 2006 However, the expression level of ABCG2 may be a useful indicator of CPT-11/SN-38 activity in lung cancer. Irinotecan 68-74 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 33-38 16542215-10 2006 However, the expression level of ABCG2 may be a useful indicator of CPT-11/SN-38 activity in lung cancer. Irinotecan 75-80 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 33-38 16517957-1 2006 BACKGROUND: Irinotecan hydrochloride (CPT-11), a topoisomerase I inhibitor highly effective for various cancers, has its dosage limited by diffuse mucosal damage with increased prostaglandin (PG) E(2). Irinotecan 12-36 topoisomerase I Zea mays 49-64 16517957-1 2006 BACKGROUND: Irinotecan hydrochloride (CPT-11), a topoisomerase I inhibitor highly effective for various cancers, has its dosage limited by diffuse mucosal damage with increased prostaglandin (PG) E(2). Irinotecan 38-44 topoisomerase I Zea mays 49-64 16465425-4 2006 MVP protein levels were enhanced in SW-620 cells after a 72 h treatment with doxorubicin (Adr), etoposide (VP-16), cis-platinum (II) diammine dichloride (CDDP) or SN-38, but not vincristine (VCR) or paclitaxel (Taxol) at their IC50 concentration. Irinotecan 163-168 major vault protein Homo sapiens 0-3 16535772-0 2006 Re: Yin MB, Li Z-R, Cao S, Durrani FA, Azrak RG, Frank C, and Rustum YM (2004) Enhanced 7-ethyl-10-hydroxycamptothecin (SN-38) lethality by methylselenocysteine is associated with Chk2 phosphorylation at threonin-68 and down-regulation of Cdc6 expression. Irinotecan 88-118 checkpoint kinase 2 Homo sapiens 180-184 16535772-0 2006 Re: Yin MB, Li Z-R, Cao S, Durrani FA, Azrak RG, Frank C, and Rustum YM (2004) Enhanced 7-ethyl-10-hydroxycamptothecin (SN-38) lethality by methylselenocysteine is associated with Chk2 phosphorylation at threonin-68 and down-regulation of Cdc6 expression. Irinotecan 88-118 cell division cycle 6 Homo sapiens 239-243 16535772-0 2006 Re: Yin MB, Li Z-R, Cao S, Durrani FA, Azrak RG, Frank C, and Rustum YM (2004) Enhanced 7-ethyl-10-hydroxycamptothecin (SN-38) lethality by methylselenocysteine is associated with Chk2 phosphorylation at threonin-68 and down-regulation of Cdc6 expression. Irinotecan 120-125 checkpoint kinase 2 Homo sapiens 180-184 16535772-0 2006 Re: Yin MB, Li Z-R, Cao S, Durrani FA, Azrak RG, Frank C, and Rustum YM (2004) Enhanced 7-ethyl-10-hydroxycamptothecin (SN-38) lethality by methylselenocysteine is associated with Chk2 phosphorylation at threonin-68 and down-regulation of Cdc6 expression. Irinotecan 120-125 cell division cycle 6 Homo sapiens 239-243 16465425-5 2006 Treatment for 48 h with Adr, VP-16 and SN-38 at their IC50 concentration also enhanced the expression of MVP mRNA. Irinotecan 39-44 major vault protein Homo sapiens 105-108 16476837-10 2006 Irinotecan and agents like gefitinib, erlotinib, and imatinib targeting the epidermal growth factor receptor and platelet-derived growth factor receptor have shown some promise in recurrent malignant glioma. Irinotecan 0-10 epidermal growth factor receptor Homo sapiens 76-108 16444269-3 2006 A cytotoxin-armed antibody reactive with one of these drug-induced surface proteins, the LY6D/E48 antigen, originally identified as the target of a monoclonal antibody reactive with squamous cell carcinomas, caused complete regression of colorectal tumor xenografts in mice treated with CPT-11, whereas either agent alone was less effective. Irinotecan 287-293 lymphocyte antigen 6 complex, locus D Mus musculus 89-93 16271446-3 2006 Irinotecan is subject to extensive metabolism by various polymorphic enzymes, including CES2 to form SN-38, members of the UGT1A subfamily, and CYP3A4 and CYP3A5, which form several pharmacologically inactive oxidation products. Irinotecan 0-10 carboxylesterase 2 Homo sapiens 88-92 16271446-3 2006 Irinotecan is subject to extensive metabolism by various polymorphic enzymes, including CES2 to form SN-38, members of the UGT1A subfamily, and CYP3A4 and CYP3A5, which form several pharmacologically inactive oxidation products. Irinotecan 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 144-150 16271446-3 2006 Irinotecan is subject to extensive metabolism by various polymorphic enzymes, including CES2 to form SN-38, members of the UGT1A subfamily, and CYP3A4 and CYP3A5, which form several pharmacologically inactive oxidation products. Irinotecan 0-10 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 155-161 16271446-3 2006 Irinotecan is subject to extensive metabolism by various polymorphic enzymes, including CES2 to form SN-38, members of the UGT1A subfamily, and CYP3A4 and CYP3A5, which form several pharmacologically inactive oxidation products. Irinotecan 101-106 carboxylesterase 2 Homo sapiens 88-92 16271446-4 2006 Elimination of irinotecan is also dependent on drug-transporting proteins, notably ABCB1 (P-glycoprotein), ABCC2 (cMOAT) and ABCG2 (BCRP), present on the bile canalicular membrane. Irinotecan 15-25 ATP binding cassette subfamily B member 1 Homo sapiens 83-88 16271446-4 2006 Elimination of irinotecan is also dependent on drug-transporting proteins, notably ABCB1 (P-glycoprotein), ABCC2 (cMOAT) and ABCG2 (BCRP), present on the bile canalicular membrane. Irinotecan 15-25 ATP binding cassette subfamily C member 2 Homo sapiens 107-112 16271446-4 2006 Elimination of irinotecan is also dependent on drug-transporting proteins, notably ABCB1 (P-glycoprotein), ABCC2 (cMOAT) and ABCG2 (BCRP), present on the bile canalicular membrane. Irinotecan 15-25 ATP binding cassette subfamily C member 2 Homo sapiens 114-119 16271446-4 2006 Elimination of irinotecan is also dependent on drug-transporting proteins, notably ABCB1 (P-glycoprotein), ABCC2 (cMOAT) and ABCG2 (BCRP), present on the bile canalicular membrane. Irinotecan 15-25 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 125-130 16271446-4 2006 Elimination of irinotecan is also dependent on drug-transporting proteins, notably ABCB1 (P-glycoprotein), ABCC2 (cMOAT) and ABCG2 (BCRP), present on the bile canalicular membrane. Irinotecan 15-25 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 132-136 15893418-5 2006 In HeLa cells, genistein inhibited CDK1 phosphorylation after irinotecan treatment. Irinotecan 62-72 cyclin dependent kinase 1 Homo sapiens 35-39 16501908-5 2006 The EGF receptor antibody cetuximab shows activity as single agent and in combination with irinotecan. Irinotecan 91-101 epidermal growth factor receptor Homo sapiens 4-16 16877259-0 2006 Pharmacogenetics of uridine diphosphoglucuronosyltransferase (UGT) 1A family members and its role in patient response to irinotecan. Irinotecan 121-131 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 20-69 16170360-6 2006 Irinotecan treatment also induced in mouse tumours derived from the p53(wt) cell lines a tetraploid G1 arrest and in those derived from the p53-deficient cell lines a transient G2/M arrest and apoptosis. Irinotecan 0-10 transformation related protein 53, pseudogene Mus musculus 68-71 16170360-6 2006 Irinotecan treatment also induced in mouse tumours derived from the p53(wt) cell lines a tetraploid G1 arrest and in those derived from the p53-deficient cell lines a transient G2/M arrest and apoptosis. Irinotecan 0-10 transformation related protein 53, pseudogene Mus musculus 140-143 16454746-3 2006 However, overexpression of ABCG2 (BCRP/MXR/ABCP) reportedly confers cancer cells resistance to SN-38, the active form of CPT-11. Irinotecan 95-100 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 27-32 16454746-3 2006 However, overexpression of ABCG2 (BCRP/MXR/ABCP) reportedly confers cancer cells resistance to SN-38, the active form of CPT-11. Irinotecan 95-100 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 34-38 16454746-3 2006 However, overexpression of ABCG2 (BCRP/MXR/ABCP) reportedly confers cancer cells resistance to SN-38, the active form of CPT-11. Irinotecan 95-100 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 39-42 16454746-3 2006 However, overexpression of ABCG2 (BCRP/MXR/ABCP) reportedly confers cancer cells resistance to SN-38, the active form of CPT-11. Irinotecan 95-100 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 43-47 16454746-3 2006 However, overexpression of ABCG2 (BCRP/MXR/ABCP) reportedly confers cancer cells resistance to SN-38, the active form of CPT-11. Irinotecan 121-127 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 27-32 16454746-3 2006 However, overexpression of ABCG2 (BCRP/MXR/ABCP) reportedly confers cancer cells resistance to SN-38, the active form of CPT-11. Irinotecan 121-127 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 34-38 16454746-3 2006 However, overexpression of ABCG2 (BCRP/MXR/ABCP) reportedly confers cancer cells resistance to SN-38, the active form of CPT-11. Irinotecan 121-127 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 39-42 16454746-3 2006 However, overexpression of ABCG2 (BCRP/MXR/ABCP) reportedly confers cancer cells resistance to SN-38, the active form of CPT-11. Irinotecan 121-127 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 43-47 17163168-9 2006 Tumors with high TS, TP, and DPD expression levels should be treated with such non-TS-directed anticancer drugs as irinotecan or oxaliplatin, or in combination with 5-FU. Irinotecan 115-125 dihydropyrimidine dehydrogenase Homo sapiens 29-32 16251201-0 2006 Phase II study of tailored chemotherapy for advanced colorectal cancer with either 5-fluouracil and leucovorin or oxaliplatin and irinotecan based on the expression of thymidylate synthase and dihydropyrimidine dehydrogenase. Irinotecan 130-140 thymidylate synthetase Homo sapiens 168-188 16251201-0 2006 Phase II study of tailored chemotherapy for advanced colorectal cancer with either 5-fluouracil and leucovorin or oxaliplatin and irinotecan based on the expression of thymidylate synthase and dihydropyrimidine dehydrogenase. Irinotecan 130-140 dihydropyrimidine dehydrogenase Homo sapiens 193-224 16877259-4 2006 SN-38 is inactivated via glucuronidation catalyzed by various hepatic and extrahepatic UGT1A isozymes. Irinotecan 0-5 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 87-92 16877259-5 2006 Although the role of the UGT1A1 *28 genetic variant has received much attention in altered toxicity upon irinotecan treatment, other UGT1A enzymes also play an important role. Irinotecan 105-115 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 25-31 16877259-5 2006 Although the role of the UGT1A1 *28 genetic variant has received much attention in altered toxicity upon irinotecan treatment, other UGT1A enzymes also play an important role. Irinotecan 105-115 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 25-30 16409125-4 2006 The objective of this study was to determine whether the chemotherapeutic agent irinotecan, by enhancing nuclear localization of YB-1, and the histone deacetylase inhibitor trichostatin A, by upregulating coxsackievirus-adenovirus receptor (CAR) expression, could augment replication of and cell lysis by adenovirus dl520 in glioblastomas in vitro. Irinotecan 80-90 Y-box binding protein 1 Homo sapiens 129-133 16409125-4 2006 The objective of this study was to determine whether the chemotherapeutic agent irinotecan, by enhancing nuclear localization of YB-1, and the histone deacetylase inhibitor trichostatin A, by upregulating coxsackievirus-adenovirus receptor (CAR) expression, could augment replication of and cell lysis by adenovirus dl520 in glioblastomas in vitro. Irinotecan 80-90 CXADR Ig-like cell adhesion molecule Homo sapiens 241-244 16409125-5 2006 We found that trichostatin A upregulated CAR expression and that irinotecan caused increased nuclear localization of YB-1 in both glioblastoma cell lines. Irinotecan 65-75 Y-box binding protein 1 Homo sapiens 117-121 17878748-1 2006 OBJECTIVES: Breast cancer resistance protein (BCRP) confers resistance to certain anticancer drugs such as mitoxantrone, topotecan and SN-38. Irinotecan 135-140 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 12-44 17878748-1 2006 OBJECTIVES: Breast cancer resistance protein (BCRP) confers resistance to certain anticancer drugs such as mitoxantrone, topotecan and SN-38. Irinotecan 135-140 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 46-50 16314881-1 2006 Common polymorphisms within the human UGT1A gene locus are associated with irinotecan and tranilast toxicity. Irinotecan 75-85 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 38-43 16267624-0 2005 Pharmacogenetics of irinotecan: a promoter polymorphism of UGT1A1 gene and severe adverse reactions to irinotecan. Irinotecan 20-30 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 59-65 15918039-6 2005 Consistent with the pharmacokinetic data, four to fivefold more CPT-11 was required to induce regressions in human Rh30 xenografts grown in esterase-deficient scid mice, as opposed to those grown in scid animals. Irinotecan 64-70 Rh blood group D antigen Homo sapiens 115-119 16257398-0 2005 Inhibition of acetylcholinesterase by the anticancer prodrug CPT-11. Irinotecan 61-67 acetylcholinesterase (Cartwright blood group) Homo sapiens 14-34 16257398-6 2005 Kinetic studies indicated that CPT-11 was primarily responsible for AChE inhibition with the 4-piperidinopiperidine moiety, the major determinant in the loss of enzyme activity. Irinotecan 31-37 acetylcholinesterase (Cartwright blood group) Homo sapiens 68-72 16367905-5 2005 PA317 cells transfected with C603S-BCRP (PA/C603S) showed either similar or only marginally lower SN-38 resistance than PA/WT cells, despite the reduced levels of BCRP dimer in these cells. Irinotecan 98-103 ATP binding cassette subfamily G member 2 (Junior blood group) Mus musculus 34-39 16367905-5 2005 PA317 cells transfected with C603S-BCRP (PA/C603S) showed either similar or only marginally lower SN-38 resistance than PA/WT cells, despite the reduced levels of BCRP dimer in these cells. Irinotecan 98-103 ATP binding cassette subfamily G member 2 (Junior blood group) Mus musculus 35-39 16367905-6 2005 Moreover, the degree of SN-38 resistance in the mutant BCRP transfectants was found to be associated with the monomer expression levels under reducing conditions. Irinotecan 24-29 ATP binding cassette subfamily G member 2 (Junior blood group) Mus musculus 55-59 16123050-0 2005 Gefitinib (Iressa) inhibits the CYP3A4-mediated formation of 7-ethyl-10-(4-amino-1-piperidino)carbonyloxycamptothecin but activates that of 7-ethyl-10-[4-N-(5-aminopentanoic acid)-1-piperidino]carbonyloxycamptothecin from irinotecan. Irinotecan 222-232 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 32-38 16267624-1 2005 This review focuses on a pharmacogenetic association between genetic polymorphism of UGT1A1 gene and severe adverse reactions to irinotecan. Irinotecan 129-139 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 85-91 16267624-4 2005 A case-control study of Japanese cancer patients revealed that those with the variant UGT1A1 alleles were at significantly higher risk of severe adverse reactions to irinotecan, suggesting that the genotyping strategy would be clinically useful. Irinotecan 166-176 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 86-92 16400742-3 2005 (2) Cetuximab, an antibody directed against the epidermal growth factor receptor (EGFR), is indicated for patients with EGF-expressing metastatic colorectal cancer, after failure of irinotecan-based chemotherapy. Irinotecan 182-192 epidermal growth factor receptor Homo sapiens 48-80 16369175-8 2005 PTK787 alone or in combination with CPT-11 reduced the phosphorylation of VEGF-R in tumor cells and tumor-associated endothelial cells, was associated with decreased microvessel density, a decreased proliferative index, and an increased apoptotic index. Irinotecan 36-42 vascular endothelial growth factor A Mus musculus 74-78 16373718-2 2005 We assessed the roles of p53, TRAIL receptors, and cellular Fas-associated death domain-like interleukin-1beta-converting enzyme inhibitory protein (c-FLIP) in regulating the cytotoxic effects of recombinant TRAIL (rTRAIL) alone and in combination with chemotherapy [5-fluorouracil (5-FU), oxaliplatin, and irinotecan] in a panel of colon cancer cell lines. Irinotecan 307-317 CASP8 and FADD like apoptosis regulator Homo sapiens 149-155 16491625-0 2005 UGT1A1 genotyping in patients undergoing treatment with irinotecan. Irinotecan 56-66 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 16095811-0 2005 The radical scavenger edaravone enhances the anti-tumor effects of CPT-11 in murine colon cancer by increasing apoptosis via inhibition of NF-kappaB. Irinotecan 67-73 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 139-148 16095811-4 2005 In vitro, SN38, the active metabolite of CPT-11, induced activation of NF-kB, the production of intracellular reactive oxygen species, the activation of caspase-3, and apoptosis in colon26 cells. Irinotecan 41-47 caspase 3 Mus musculus 153-162 16399432-4 2005 Patients who have failed first-line therapy have benefited from 5-FU, leucovorin, and irinotecan (IFL), 5-FU, leucovorin, and oxaliplatin (FOLFOX), and 5-FU, leucovorin, and irinotecan (FOLFIRI) in the second-line treatment setting. Irinotecan 86-96 interferon alpha 1 Homo sapiens 98-101 16222152-0 2005 The epidermal growth factor receptor tyrosine kinase inhibitor gefitinib sensitizes colon cancer cells to irinotecan. Irinotecan 106-116 epidermal growth factor receptor Homo sapiens 4-36 16222152-3 2005 We investigated the effects of the EGFR tyrosine kinase inhibitor gefitinib on cellular determinants of irinotecan resistance in human colon cancer cells. Irinotecan 104-114 epidermal growth factor receptor Homo sapiens 35-39 15947933-8 2005 CPT-11 80 mg/m(2) produced a mean increase in S-phase by IHC for cyclin A of 137%. Irinotecan 0-6 cyclin A2 Homo sapiens 65-73 16132345-3 2005 P-glycoprotein, breast cancer resistance protein, MRP1, and MRP2 have been implicated in resistance to various CPTs including CPT-11 (irinotecan), SN-38 (the active metabolite of CPT-11), and topotecan. Irinotecan 126-132 ATP binding cassette subfamily B member 1 Homo sapiens 0-14 16132345-12 2005 In addition, the accumulation of CPT-11 and SN-38 over 120 min in MRP4/HepG2 cells was significantly reduced compared to V/HepG2 cells, whereas the addition of celecoxib, MK571, or BSO significantly increased their accumulation in MRP4/HepG2 cells. Irinotecan 33-39 ATP binding cassette subfamily C member 4 Homo sapiens 66-70 16132345-12 2005 In addition, the accumulation of CPT-11 and SN-38 over 120 min in MRP4/HepG2 cells was significantly reduced compared to V/HepG2 cells, whereas the addition of celecoxib, MK571, or BSO significantly increased their accumulation in MRP4/HepG2 cells. Irinotecan 33-39 ATP binding cassette subfamily C member 4 Homo sapiens 231-235 16132345-12 2005 In addition, the accumulation of CPT-11 and SN-38 over 120 min in MRP4/HepG2 cells was significantly reduced compared to V/HepG2 cells, whereas the addition of celecoxib, MK571, or BSO significantly increased their accumulation in MRP4/HepG2 cells. Irinotecan 44-49 ATP binding cassette subfamily C member 4 Homo sapiens 66-70 16270098-4 2005 For the treatment of metastatic colorectal cancer, improved response rates and prolonged survival have been reported when irinotecan or oxaliplatin was added to 5-FU/FA; a further increase in efficacy was shown when bevacizumab, an antibody to vascular endothelial growth factor, was added to chemotherapy. Irinotecan 122-132 vascular endothelial growth factor A Homo sapiens 244-278 16132345-17 2005 CPT-11 and SN-38 are substrates for MRP4. Irinotecan 0-6 ATP binding cassette subfamily C member 4 Homo sapiens 36-40 16132345-17 2005 CPT-11 and SN-38 are substrates for MRP4. Irinotecan 11-16 ATP binding cassette subfamily C member 4 Homo sapiens 36-40 16132345-12 2005 In addition, the accumulation of CPT-11 and SN-38 over 120 min in MRP4/HepG2 cells was significantly reduced compared to V/HepG2 cells, whereas the addition of celecoxib, MK571, or BSO significantly increased their accumulation in MRP4/HepG2 cells. Irinotecan 44-49 ATP binding cassette subfamily C member 4 Homo sapiens 231-235 16132345-3 2005 P-glycoprotein, breast cancer resistance protein, MRP1, and MRP2 have been implicated in resistance to various CPTs including CPT-11 (irinotecan), SN-38 (the active metabolite of CPT-11), and topotecan. Irinotecan 126-132 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 16-48 16132345-16 2005 CONCLUSIONS: Human MRP4 rendered significant resistance to cyclophosphamide, CPT, CPT-11, SN-38, rubitecan, and 10-OH-CPT. Irinotecan 82-88 ATP binding cassette subfamily C member 4 Homo sapiens 19-23 16132345-16 2005 CONCLUSIONS: Human MRP4 rendered significant resistance to cyclophosphamide, CPT, CPT-11, SN-38, rubitecan, and 10-OH-CPT. Irinotecan 90-95 ATP binding cassette subfamily C member 4 Homo sapiens 19-23 16132345-3 2005 P-glycoprotein, breast cancer resistance protein, MRP1, and MRP2 have been implicated in resistance to various CPTs including CPT-11 (irinotecan), SN-38 (the active metabolite of CPT-11), and topotecan. Irinotecan 126-132 ATP binding cassette subfamily C member 1 Homo sapiens 50-54 16132345-3 2005 P-glycoprotein, breast cancer resistance protein, MRP1, and MRP2 have been implicated in resistance to various CPTs including CPT-11 (irinotecan), SN-38 (the active metabolite of CPT-11), and topotecan. Irinotecan 126-132 ATP binding cassette subfamily C member 2 Homo sapiens 60-64 16132345-3 2005 P-glycoprotein, breast cancer resistance protein, MRP1, and MRP2 have been implicated in resistance to various CPTs including CPT-11 (irinotecan), SN-38 (the active metabolite of CPT-11), and topotecan. Irinotecan 134-144 ATP binding cassette subfamily B member 1 Homo sapiens 0-14 16132345-3 2005 P-glycoprotein, breast cancer resistance protein, MRP1, and MRP2 have been implicated in resistance to various CPTs including CPT-11 (irinotecan), SN-38 (the active metabolite of CPT-11), and topotecan. Irinotecan 134-144 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 16-48 16132345-3 2005 P-glycoprotein, breast cancer resistance protein, MRP1, and MRP2 have been implicated in resistance to various CPTs including CPT-11 (irinotecan), SN-38 (the active metabolite of CPT-11), and topotecan. Irinotecan 134-144 ATP binding cassette subfamily C member 1 Homo sapiens 50-54 16132345-3 2005 P-glycoprotein, breast cancer resistance protein, MRP1, and MRP2 have been implicated in resistance to various CPTs including CPT-11 (irinotecan), SN-38 (the active metabolite of CPT-11), and topotecan. Irinotecan 134-144 ATP binding cassette subfamily C member 2 Homo sapiens 60-64 16132345-3 2005 P-glycoprotein, breast cancer resistance protein, MRP1, and MRP2 have been implicated in resistance to various CPTs including CPT-11 (irinotecan), SN-38 (the active metabolite of CPT-11), and topotecan. Irinotecan 147-152 ATP binding cassette subfamily B member 1 Homo sapiens 0-14 16132345-3 2005 P-glycoprotein, breast cancer resistance protein, MRP1, and MRP2 have been implicated in resistance to various CPTs including CPT-11 (irinotecan), SN-38 (the active metabolite of CPT-11), and topotecan. Irinotecan 147-152 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 16-48 16132345-3 2005 P-glycoprotein, breast cancer resistance protein, MRP1, and MRP2 have been implicated in resistance to various CPTs including CPT-11 (irinotecan), SN-38 (the active metabolite of CPT-11), and topotecan. Irinotecan 147-152 ATP binding cassette subfamily C member 1 Homo sapiens 50-54 16132345-3 2005 P-glycoprotein, breast cancer resistance protein, MRP1, and MRP2 have been implicated in resistance to various CPTs including CPT-11 (irinotecan), SN-38 (the active metabolite of CPT-11), and topotecan. Irinotecan 147-152 ATP binding cassette subfamily C member 2 Homo sapiens 60-64 16132345-3 2005 P-glycoprotein, breast cancer resistance protein, MRP1, and MRP2 have been implicated in resistance to various CPTs including CPT-11 (irinotecan), SN-38 (the active metabolite of CPT-11), and topotecan. Irinotecan 179-185 ATP binding cassette subfamily B member 1 Homo sapiens 0-14 16132345-3 2005 P-glycoprotein, breast cancer resistance protein, MRP1, and MRP2 have been implicated in resistance to various CPTs including CPT-11 (irinotecan), SN-38 (the active metabolite of CPT-11), and topotecan. Irinotecan 179-185 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 16-48 16132345-3 2005 P-glycoprotein, breast cancer resistance protein, MRP1, and MRP2 have been implicated in resistance to various CPTs including CPT-11 (irinotecan), SN-38 (the active metabolite of CPT-11), and topotecan. Irinotecan 179-185 ATP binding cassette subfamily C member 1 Homo sapiens 50-54 16132345-3 2005 P-glycoprotein, breast cancer resistance protein, MRP1, and MRP2 have been implicated in resistance to various CPTs including CPT-11 (irinotecan), SN-38 (the active metabolite of CPT-11), and topotecan. Irinotecan 179-185 ATP binding cassette subfamily C member 2 Homo sapiens 60-64 16098482-0 2005 MRP1 mutated in the L0 region transports SN-38 but not leukotriene C4 or estradiol-17 (beta-D-glucuronate). Irinotecan 41-46 ATP binding cassette subfamily B member 1 Homo sapiens 0-4 16315940-5 2005 A weekly CPT-11 infusion therapy was effective to the recurrent skin and soft tissue lesions (CR and PR), and the tumor markers (CEA and CA15-3) markedly declined. Irinotecan 9-15 mucin 1, cell surface associated Homo sapiens 137-143 16204068-3 2005 In contrast, irinotecan (CPT-11) and tomudex sensitized p53 WT, mutant, and null cells to Fas-mediated cell death. Irinotecan 13-23 tumor protein p53 Homo sapiens 56-59 16204068-3 2005 In contrast, irinotecan (CPT-11) and tomudex sensitized p53 WT, mutant, and null cells to Fas-mediated cell death. Irinotecan 25-31 tumor protein p53 Homo sapiens 56-59 16204068-4 2005 Furthermore, CPT-11 and tomudex, but not 5-FU or oxaliplatin, up-regulated Fas cell surface expression in a p53-independent manner. Irinotecan 13-19 tumor protein p53 Homo sapiens 108-111 16204068-5 2005 In addition, increased Fas cell surface expression in p53 mutant and null cell lines in response to CPT-11 and tomudex was accompanied by only a slight increase in total Fas mRNA and protein expression, suggesting that these agents trigger p53-independent trafficking of Fas to the plasma membrane. Irinotecan 100-106 tumor protein p53 Homo sapiens 54-57 16204068-6 2005 Treatment with CPT-11 or tomudex induced STAT1 phosphorylation (Ser727) in the p53-null HCT116 cell line but not the p53 WT cell line. Irinotecan 15-21 signal transducer and activator of transcription 1 Homo sapiens 41-46 16204068-6 2005 Treatment with CPT-11 or tomudex induced STAT1 phosphorylation (Ser727) in the p53-null HCT116 cell line but not the p53 WT cell line. Irinotecan 15-21 tumor protein p53 Homo sapiens 79-82 16204068-7 2005 Furthermore, STAT1-targeted small interfering RNA (siRNA) inhibited up-regulation of Fas cell surface expression in response to CPT-11 and tomudex in these cells. Irinotecan 128-134 signal transducer and activator of transcription 1 Homo sapiens 13-18 16204068-9 2005 This suggests that STAT1 regulates expression of gene(s) involved in cell surface trafficking of Fas in response to CPT-11 or tomudex. Irinotecan 116-122 signal transducer and activator of transcription 1 Homo sapiens 19-24 16204068-10 2005 We conclude that CPT-11 and tomudex may be more effective than 5-FU and oxaliplatin in the treatment of p53 mutant colorectal cancer tumors by sensitizing them to Fas-mediated apoptosis in a STAT1-dependent manner. Irinotecan 17-23 tumor protein p53 Homo sapiens 104-107 16204068-10 2005 We conclude that CPT-11 and tomudex may be more effective than 5-FU and oxaliplatin in the treatment of p53 mutant colorectal cancer tumors by sensitizing them to Fas-mediated apoptosis in a STAT1-dependent manner. Irinotecan 17-23 signal transducer and activator of transcription 1 Homo sapiens 191-196 16203781-1 2005 PURPOSE: Irinotecan (CPT11) is a prodrug activated in humans mainly by carboxylesterase 2 (CES2) generating the SN38 metabolite responsible for the drug efficacy and toxicity. Irinotecan 9-19 carboxylesterase 2 Homo sapiens 71-89 16203781-1 2005 PURPOSE: Irinotecan (CPT11) is a prodrug activated in humans mainly by carboxylesterase 2 (CES2) generating the SN38 metabolite responsible for the drug efficacy and toxicity. Irinotecan 9-19 carboxylesterase 2 Homo sapiens 91-95 16203781-1 2005 PURPOSE: Irinotecan (CPT11) is a prodrug activated in humans mainly by carboxylesterase 2 (CES2) generating the SN38 metabolite responsible for the drug efficacy and toxicity. Irinotecan 21-26 carboxylesterase 2 Homo sapiens 71-89 16203781-1 2005 PURPOSE: Irinotecan (CPT11) is a prodrug activated in humans mainly by carboxylesterase 2 (CES2) generating the SN38 metabolite responsible for the drug efficacy and toxicity. Irinotecan 21-26 carboxylesterase 2 Homo sapiens 91-95 16251801-3 2005 By treating fresh human colonic explants with 5-Fluorouracil (200 microg/ml), CPT-11 (100 microg/ml) and/or TRAIL (100 ng/ml) we readily detected a signal in situ using FITC-VAD-FMK at different time points, whereas labeling of colonic explants with EGFP-conjugated Annexin V proved less specific. Irinotecan 78-84 annexin A5 Homo sapiens 266-275 16166450-3 2005 A recent study found that coadministration of irinotecan and ketoconazole led to a significant increase in the formation of SN-38 (7-ethyl-10-hydroxycamptothecine), an UGT1A substrate. Irinotecan 46-56 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 168-173 16166450-3 2005 A recent study found that coadministration of irinotecan and ketoconazole led to a significant increase in the formation of SN-38 (7-ethyl-10-hydroxycamptothecine), an UGT1A substrate. Irinotecan 124-129 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 168-173 16166450-3 2005 A recent study found that coadministration of irinotecan and ketoconazole led to a significant increase in the formation of SN-38 (7-ethyl-10-hydroxycamptothecine), an UGT1A substrate. Irinotecan 131-162 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 168-173 16166450-5 2005 EXPERIMENTAL DESIGN: SN-38 glucuronidation activities were determined by measuring the rate of SN-38 glucuronide (SN-38G) formation using pooled human liver microsomes and cDNA-expressed UGT1A isoforms (1A1, 1A7 and 1A9) in the presence of ketoconazole. Irinotecan 21-26 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 187-192 16166450-11 2005 CONCLUSIONS: These results show that ketoconazole is a potent UGT1A1 inhibitor, which seems the basis for increased exposure to SN-38 when coadministered with irinotecan. Irinotecan 128-133 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 62-68 16166450-11 2005 CONCLUSIONS: These results show that ketoconazole is a potent UGT1A1 inhibitor, which seems the basis for increased exposure to SN-38 when coadministered with irinotecan. Irinotecan 159-169 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 62-68 16144935-10 2005 In conclusion, low SMAD4 tumor levels identified a subset of patients with poor prognosis following surgery and 5-FU-based adjuvant therapy; therefore, these patients could be good candidates to receive combined treatment with additional chemotherapeutic agents such as CPT-11 and/or oxaliplatin. Irinotecan 270-276 SMAD family member 4 Homo sapiens 19-24 15828025-5 2005 The main aim of this review is to provide an overview of the known polymorphisms present in the genes which codify for factors (thymidylate synthase dihydropyrimidine dehydrogenase, uridine diphosphate (UDP)-glucuronosyl-transferase 1A1, enzymes implicated in DNA repair) involved in the action mechanisms of the drugs now utilized in chemotherapeutic treatment of colorectal carcinoma, such as fluoropyrimidines, irinotecan, and platinum agents. Irinotecan 414-424 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 182-236 16184937-0 2005 [A case of colon cancer with liver and lung metastases-efficacy of CPT-11/5-FU/l-LV (IFL) as part of ambulatory treatment]. Irinotecan 67-73 interferon alpha 1 Homo sapiens 85-88 16101140-2 2005 PATIENTS AND METHODS: The impact of gemcitabine, irinotecan and oxaliplatin + 5-FU upon the serum markers vascular endothelial growth factor (VEGF) (pro-angiogenic) and IFN-gamma-inducible protein (IP)-10 (anti-angiogenic) was evaluated by ELISA in locally advanced and/or metastatic cancer versus clinical efficacy and survival. Irinotecan 49-59 vascular endothelial growth factor A Homo sapiens 106-140 16187025-4 2005 Irinotecan is metabolized by carboxylesterase to form an active SN-38, which is further conjugated and detoxified by UDP-glucuronosyltransferase (UGT) to yield its beta-glucuronide. Irinotecan 0-10 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 117-144 16187025-4 2005 Irinotecan is metabolized by carboxylesterase to form an active SN-38, which is further conjugated and detoxified by UDP-glucuronosyltransferase (UGT) to yield its beta-glucuronide. Irinotecan 0-10 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 146-149 16187025-4 2005 Irinotecan is metabolized by carboxylesterase to form an active SN-38, which is further conjugated and detoxified by UDP-glucuronosyltransferase (UGT) to yield its beta-glucuronide. Irinotecan 64-69 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 117-144 16187025-4 2005 Irinotecan is metabolized by carboxylesterase to form an active SN-38, which is further conjugated and detoxified by UDP-glucuronosyltransferase (UGT) to yield its beta-glucuronide. Irinotecan 64-69 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 146-149 16187025-5 2005 The polymorphic UGT isoenzyme, UGT1A1 has genetic variants which decrease in SN-38 glucuronidating capacity and could help predict irinotecan-associated toxicity. Irinotecan 77-82 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 16-19 16187025-5 2005 The polymorphic UGT isoenzyme, UGT1A1 has genetic variants which decrease in SN-38 glucuronidating capacity and could help predict irinotecan-associated toxicity. Irinotecan 77-82 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 31-37 16187025-5 2005 The polymorphic UGT isoenzyme, UGT1A1 has genetic variants which decrease in SN-38 glucuronidating capacity and could help predict irinotecan-associated toxicity. Irinotecan 131-141 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 16-19 16187025-5 2005 The polymorphic UGT isoenzyme, UGT1A1 has genetic variants which decrease in SN-38 glucuronidating capacity and could help predict irinotecan-associated toxicity. Irinotecan 131-141 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 31-37 16187025-7 2005 We suggest that irinotecan offers an effective treatment option for children with recurrent brain stem glioma and other genetic variants except UGT1A1 may be a risk factor for irinotecan-induced toxicity. Irinotecan 176-186 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 144-150 16010439-0 2005 Increased anticancer activity of the thymidylate synthase inhibitor BGC9331 combined with the topoisomerase I inhibitor SN-38 in human colorectal and breast cancer cells: induction of apoptosis and ROCK cleavage through caspase-3-dependent and -independent mechanisms. Irinotecan 120-125 caspase 3 Homo sapiens 220-229 16077970-2 2005 We examined TS expression in tumours from 86 patients with advanced colorectal cancer who received one of two chemotherapy regimes (either irinotecan alone or irinotecan and 5-flurouracil with folinic acid). Irinotecan 139-149 thymidylate synthetase Homo sapiens 12-14 16077970-2 2005 We examined TS expression in tumours from 86 patients with advanced colorectal cancer who received one of two chemotherapy regimes (either irinotecan alone or irinotecan and 5-flurouracil with folinic acid). Irinotecan 159-169 thymidylate synthetase Homo sapiens 12-14 16077970-6 2005 A significant association was found between tumour TS expression and response to treatment with 5-FU plus FA with irinotecan (p=0.05). Irinotecan 114-124 thymidylate synthetase Homo sapiens 51-53 16108826-1 2005 Breast cancer resistance protein (BCRP) is a half-molecule ATP-binding cassette transporter that forms a functional homodimer and pumps out various anticancer agents, such as 7-ethyl-10-hydroxycamptothecin, topotecan, mitoxantrone and flavopiridol, from cells. Irinotecan 175-205 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 0-32 16108826-1 2005 Breast cancer resistance protein (BCRP) is a half-molecule ATP-binding cassette transporter that forms a functional homodimer and pumps out various anticancer agents, such as 7-ethyl-10-hydroxycamptothecin, topotecan, mitoxantrone and flavopiridol, from cells. Irinotecan 175-205 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 34-38 16108826-1 2005 Breast cancer resistance protein (BCRP) is a half-molecule ATP-binding cassette transporter that forms a functional homodimer and pumps out various anticancer agents, such as 7-ethyl-10-hydroxycamptothecin, topotecan, mitoxantrone and flavopiridol, from cells. Irinotecan 175-205 ATP binding cassette subfamily A member 4 Homo sapiens 59-91 16103093-3 2005 The combination IFN-beta and irinotecan (CPT-11) cooperatively inhibits cell growth and IRF-5 synergizes with it to further promote apoptosis. Irinotecan 29-39 interferon regulatory factor 5 Homo sapiens 88-93 16103093-3 2005 The combination IFN-beta and irinotecan (CPT-11) cooperatively inhibits cell growth and IRF-5 synergizes with it to further promote apoptosis. Irinotecan 41-47 interferon beta 1 Homo sapiens 16-24 15838659-3 2005 In 4-day cytotoxicity assays with mitoxantrone, topotecan, SN-38 or diflomotecan, cells transfected with wild-type R482 ABCG2 showed IC50 values up to 1.2-fold to 5-fold higher than cells expressing comparable levels of Q141K ABCG2, suggesting that the Q141K SNP affects drug transport. Irinotecan 59-64 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 120-125 16010439-5 2005 Treatment of HT-29 cells with either BGC9331 or SN-38 increased caspase-3 activity and the percentage of apoptotic cells from 3 to 13%. Irinotecan 48-53 caspase 3 Homo sapiens 64-73 16022921-1 2005 Irinotecan is a topoisomerase I inhibitor that is highly active against small cell lung cancer (SCLC). Irinotecan 0-10 SCLC1 Homo sapiens 96-100 16022921-11 2005 This irinotecan and etoposide regimen is active against ED-SCLC with relatively mild toxicity. Irinotecan 5-15 SCLC1 Homo sapiens 59-63 15944780-4 2005 The tumor weights with MTD of CPM or CPT-11 in combination with anti-IL-6 antibody treatment, which decreases serum IL-6 level and improves cachexia status, were significantly smaller than those in the MTD treatment-alone group with clone 20, but not with clone 5. Irinotecan 37-43 interleukin 6 Mus musculus 116-120 15833930-4 2005 The stability of SN-38 lactone and carboxylate in incubation mixtures of microsomes and UDP-glucuronosyltransferase (UGT) isoforms was used to determine optimal incubation times. Irinotecan 17-30 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 88-115 16104527-0 2005 [Assessment of immunotoxicity of irinotecan determined by the novel method, by which productivity of TNF-alpha from whole blood is stimulated by lipopolysaccharide]. Irinotecan 33-43 tumor necrosis factor Homo sapiens 101-110 15772291-4 2005 Previous studies have indicated that CPT-11 can inhibit acetylcholinesterase (AChE), and here, we provide a detailed analysis of the inhibition of AChE by CPT-11 and by structural analogs. Irinotecan 37-43 acetylcholinesterase (Cartwright blood group) Homo sapiens 78-82 15846114-6 2005 In contrast, doxorubicin and paclitaxel caused no up-regulation in vitro, while irinotecan caused inhibition of COX-2 (2.7-fold decrease, p<0.01). Irinotecan 80-90 mitochondrially encoded cytochrome c oxidase II Homo sapiens 112-117 15984515-2 2005 The standard chemotherapy for extensive SCLC in Japan is the combination of irinotecan and cisplatin. Irinotecan 76-86 SCLC1 Homo sapiens 40-44 15902309-3 2005 We were able to achieve restoration of their radiosensitivity and sensitivity to 5-fluorouracil and irinotecan by reexpression of SPARC in tumor xenografts. Irinotecan 100-110 secreted protein acidic and cysteine rich Homo sapiens 130-135 15723219-2 2005 In the experiments described here using AGS gastric cancer cells, SN38 (the active metabolite of CPT-11) induced tyrosine phosphorylation of EGFR within 5 min, and this was followed by the induction of transcripts and/or proteins of heparin-binding EGF-like growth factor, amphiregulin, transforming growth factor-alpha, and interlukin-8 (IL-8). Irinotecan 97-103 epidermal growth factor receptor Homo sapiens 141-145 15930350-8 2005 Patients with UDP-glucuronosyltransferase (UGT1A1; TA)6/6 promoter genotype had a lower ratio of free to glucuronide form of SN-38 than in patients with >/=1 (TA)7 allele. Irinotecan 125-130 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 43-49 15956246-7 2005 Many gene expression changes observed following in vitro SN-38 exposure were also seen following in vivo administration of 10 or 15 mg/m(2) CPT-11; notably, proapoptotic changes included reduced transcription of survivin pathway-associated genes and increased transcription of death receptor 5. Irinotecan 57-62 TNF receptor superfamily member 10b Homo sapiens 277-293 15956246-7 2005 Many gene expression changes observed following in vitro SN-38 exposure were also seen following in vivo administration of 10 or 15 mg/m(2) CPT-11; notably, proapoptotic changes included reduced transcription of survivin pathway-associated genes and increased transcription of death receptor 5. Irinotecan 140-146 TNF receptor superfamily member 10b Homo sapiens 277-293 15772291-4 2005 Previous studies have indicated that CPT-11 can inhibit acetylcholinesterase (AChE), and here, we provide a detailed analysis of the inhibition of AChE by CPT-11 and by structural analogs. Irinotecan 37-43 acetylcholinesterase (Cartwright blood group) Homo sapiens 147-151 15772291-4 2005 Previous studies have indicated that CPT-11 can inhibit acetylcholinesterase (AChE), and here, we provide a detailed analysis of the inhibition of AChE by CPT-11 and by structural analogs. Irinotecan 155-161 acetylcholinesterase (Cartwright blood group) Homo sapiens 147-151 15772291-5 2005 These studies demonstrate that the terminal dipiperidino moiety in CPT-11 plays a major role in enzyme inhibition, and this has been confirmed by X-ray crystallographic studies of a complex of the drug with Torpedo californica AChE. Irinotecan 67-73 acetylcholinesterase (Cartwright blood group) Homo sapiens 227-231 15772291-7 2005 The 3D structure of the CPT-11/AChE complex also permits modeling of CPT-11 complexed with mammalian butyrylcholinesterase and carboxylesterase, both of which are known to hydrolyze the drug to the active metabolite. Irinotecan 24-30 acetylcholinesterase (Cartwright blood group) Homo sapiens 31-35 15772291-7 2005 The 3D structure of the CPT-11/AChE complex also permits modeling of CPT-11 complexed with mammalian butyrylcholinesterase and carboxylesterase, both of which are known to hydrolyze the drug to the active metabolite. Irinotecan 69-75 acetylcholinesterase (Cartwright blood group) Homo sapiens 31-35 15772291-8 2005 Overall, the results presented here clarify the mechanism of AChE inhibition by CPT-11 and detail the interaction of the drug with the protein. Irinotecan 80-86 acetylcholinesterase (Cartwright blood group) Homo sapiens 61-65 15948034-1 2005 CPT-11 is a DNA topoisomerase I inhibitor for the therapy of colorectal cancer, whereas St. John"s Wort (Hypericum perforatum, SJW) is a widely used herbal anti-depressant. Irinotecan 0-6 DNA topoisomerase I Rattus norvegicus 12-31 15867226-3 2005 Drg1 expression is also associated with resistance to irinotecan therapy in preclinical colorectal cancer models. Irinotecan 54-64 developmentally regulated GTP binding protein 1 Homo sapiens 0-4 15886469-5 2005 However, in HT-29 cells, the CPT-11/5-FU combination enhanced Mn-SOD activity when compared to cells treated with CPT-11 alone. Irinotecan 29-35 superoxide dismutase 2 Homo sapiens 62-68 15886469-5 2005 However, in HT-29 cells, the CPT-11/5-FU combination enhanced Mn-SOD activity when compared to cells treated with CPT-11 alone. Irinotecan 114-120 superoxide dismutase 2 Homo sapiens 62-68 15886469-7 2005 CONCLUSION: Treatment with the CPT-11/5-FU combination may promote in HT-29 cell apoptosis by enhancing Mn-SOD activity. Irinotecan 31-37 superoxide dismutase 2 Homo sapiens 104-110 15867367-7 2005 AEE788 alone or in combination with CPT-11 inhibited pEGFR, pVEGFR, and phosphorylated Akt expression on tumor-associated endothelial cells as well as on tumor cells. Irinotecan 36-42 thymoma viral proto-oncogene 1 Mus musculus 87-90 15867226-7 2005 We correlated expression of Drg1 to numerous clinical and tumor related variables and to patient outcomes, including a subset of patients who recurred and received irinotecan-based therapy. Irinotecan 164-174 developmentally regulated GTP binding protein 1 Homo sapiens 28-32 15867226-10 2005 High Drg1 expression suggested irinotecan resistance (P = 0.07). Irinotecan 31-41 developmentally regulated GTP binding protein 1 Homo sapiens 5-9 15867226-12 2005 High Drg1 may also be associated with relative resistance to irinotecan. Irinotecan 61-71 developmentally regulated GTP binding protein 1 Homo sapiens 5-9 15867226-5 2005 We performed this study to evaluate the role of Drg1 in a large cohort of patients with metastatic colorectal cancer who were irinotecan naive. Irinotecan 126-136 developmentally regulated GTP binding protein 1 Homo sapiens 48-52 15716465-10 2005 We identified a novel reduced-function TATA box SNP of the UGT1A7 gene that catalyzes irinotecan metabolite detoxification. Irinotecan 86-96 UDP glucuronosyltransferase family 1 member A7 Homo sapiens 59-65 15809721-0 2005 Blocking of PI3K/Akt pathway enhances apoptosis induced by SN-38, an active form of CPT-11, in human hepatoma cells. Irinotecan 59-64 AKT serine/threonine kinase 1 Homo sapiens 17-20 15809721-0 2005 Blocking of PI3K/Akt pathway enhances apoptosis induced by SN-38, an active form of CPT-11, in human hepatoma cells. Irinotecan 84-90 AKT serine/threonine kinase 1 Homo sapiens 17-20 15809721-3 2005 CPT-11, a derivative of camptothecin, works as type-I DNA topoisomerase inhibitor and showed a major objective response rate in patients with metastatic colorectal cancer. Irinotecan 0-6 DNA topoisomerase I Homo sapiens 47-71 15809721-4 2005 In this study, the mechanism underlying chemo-resistance to SN-38, an active form of CPT-11, in HCC was investigated in relation to anti-apoptotic pathways NF-kappaB and PI3K/Akt. Irinotecan 60-65 nuclear factor kappa B subunit 1 Homo sapiens 156-165 15809721-4 2005 In this study, the mechanism underlying chemo-resistance to SN-38, an active form of CPT-11, in HCC was investigated in relation to anti-apoptotic pathways NF-kappaB and PI3K/Akt. Irinotecan 60-65 AKT serine/threonine kinase 1 Homo sapiens 175-178 15809721-4 2005 In this study, the mechanism underlying chemo-resistance to SN-38, an active form of CPT-11, in HCC was investigated in relation to anti-apoptotic pathways NF-kappaB and PI3K/Akt. Irinotecan 85-91 nuclear factor kappa B subunit 1 Homo sapiens 156-165 15809721-4 2005 In this study, the mechanism underlying chemo-resistance to SN-38, an active form of CPT-11, in HCC was investigated in relation to anti-apoptotic pathways NF-kappaB and PI3K/Akt. Irinotecan 85-91 AKT serine/threonine kinase 1 Homo sapiens 175-178 15809721-6 2005 NF-kappaB was constitutively activated in Hep3B, and SN-38 further enhanced the nuclear translocation of NF-kappaB. Irinotecan 53-58 nuclear factor kappa B subunit 1 Homo sapiens 105-114 15809721-8 2005 On the other hand, SN-38 phosphorylated Akt and pretreatment with PI3K inhibitors increased SN-38-induced apoptosis, indicating that resistance to SN-38 in Hep3B occurs partly through the PI3K/Akt not the NF-kappaB pathway. Irinotecan 19-24 AKT serine/threonine kinase 1 Homo sapiens 40-43 15809721-8 2005 On the other hand, SN-38 phosphorylated Akt and pretreatment with PI3K inhibitors increased SN-38-induced apoptosis, indicating that resistance to SN-38 in Hep3B occurs partly through the PI3K/Akt not the NF-kappaB pathway. Irinotecan 92-97 AKT serine/threonine kinase 1 Homo sapiens 193-196 15809721-8 2005 On the other hand, SN-38 phosphorylated Akt and pretreatment with PI3K inhibitors increased SN-38-induced apoptosis, indicating that resistance to SN-38 in Hep3B occurs partly through the PI3K/Akt not the NF-kappaB pathway. Irinotecan 92-97 nuclear factor kappa B subunit 1 Homo sapiens 205-214 15809721-8 2005 On the other hand, SN-38 phosphorylated Akt and pretreatment with PI3K inhibitors increased SN-38-induced apoptosis, indicating that resistance to SN-38 in Hep3B occurs partly through the PI3K/Akt not the NF-kappaB pathway. Irinotecan 92-97 AKT serine/threonine kinase 1 Homo sapiens 193-196 15809721-8 2005 On the other hand, SN-38 phosphorylated Akt and pretreatment with PI3K inhibitors increased SN-38-induced apoptosis, indicating that resistance to SN-38 in Hep3B occurs partly through the PI3K/Akt not the NF-kappaB pathway. Irinotecan 92-97 nuclear factor kappa B subunit 1 Homo sapiens 205-214 15858133-3 2005 A heterozygous G > A transition at the 5" splicing donor consensus sequence in intron 14 leading to exon 14 skipping (IVS14+1 G > A, DPYD*2A) with partial loss of enzyme activity may be partly responsible for 5FU induced toxicity, whereas irinotecan associated toxicity may in part be explained by an aberrant UGT1A1 promoter (TA)(n) genotype underlying Gilbert"s syndrome with reduced liver glucuronidation activity. Irinotecan 245-255 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 316-322 15716465-2 2005 UGT1A1*28 is a functional UGT promoter polymorphism associated with Gilbert"s disease and severe irinotecan toxicity, which also occurs in the absence of UGT1A1*28. Irinotecan 97-107 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 15716465-2 2005 UGT1A1*28 is a functional UGT promoter polymorphism associated with Gilbert"s disease and severe irinotecan toxicity, which also occurs in the absence of UGT1A1*28. Irinotecan 97-107 beta-1,3-glucuronyltransferase 2 Homo sapiens 0-3 15716465-3 2005 The aim of this study was to identify and characterize UGT promoter variants relevant for irinotecan detoxification. Irinotecan 90-100 beta-1,3-glucuronyltransferase 2 Homo sapiens 55-58 15716465-11 2005 Its association with variants of the UGT1A1 promoter and UGT1A7 gene may influence irinotecan metabolism. Irinotecan 83-93 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 37-43 15716465-11 2005 Its association with variants of the UGT1A1 promoter and UGT1A7 gene may influence irinotecan metabolism. Irinotecan 83-93 UDP glucuronosyltransferase family 1 member A7 Homo sapiens 57-63 15864130-2 2005 SN-38, the active metabolite of the anticancer agent irinotecan, is metabolized by both UGT1A1 and UGT1A9. Irinotecan 0-5 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 88-94 15864130-2 2005 SN-38, the active metabolite of the anticancer agent irinotecan, is metabolized by both UGT1A1 and UGT1A9. Irinotecan 0-5 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 99-105 15864130-2 2005 SN-38, the active metabolite of the anticancer agent irinotecan, is metabolized by both UGT1A1 and UGT1A9. Irinotecan 53-63 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 88-94 15864130-2 2005 SN-38, the active metabolite of the anticancer agent irinotecan, is metabolized by both UGT1A1 and UGT1A9. Irinotecan 53-63 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 99-105 15864130-9 2005 Common UGT1A9-UGT1A1 diplotypes were defined, and a difference was observed across the SN-38 glucuronidation rates in Caucasian livers stratified by diplotypes. Irinotecan 87-92 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 7-13 15864130-11 2005 If the functional effect of these haplotypes can be confirmed, this haplotypic information would be applicable to the correct design of prospective clinical studies of irinotecan, as well as of other drugs primarily metabolized by both UGT1A1 and UGT1A9. Irinotecan 168-178 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 247-253 15699047-5 2005 Colony forming assays demonstrated that down-regulation of ATR or Chk1 sensitized cells to SN-38 and camptothecin. Irinotecan 91-96 ATR serine/threonine kinase Homo sapiens 59-62 15897249-10 2005 In conclusion, the significant increase in the cure rate after methylselenocysteine/CPT-11 could be related to increased drug delivery into both tumors (CES1), reduced resistance to SN-38 (ABCC1 and DRG1) in FaDu, and induced Fas ligand apoptosis (TNFSF6) in A253. Irinotecan 84-90 carboxylesterase 1 Homo sapiens 153-157 15897249-10 2005 In conclusion, the significant increase in the cure rate after methylselenocysteine/CPT-11 could be related to increased drug delivery into both tumors (CES1), reduced resistance to SN-38 (ABCC1 and DRG1) in FaDu, and induced Fas ligand apoptosis (TNFSF6) in A253. Irinotecan 84-90 ATP binding cassette subfamily C member 1 Homo sapiens 189-194 15897249-10 2005 In conclusion, the significant increase in the cure rate after methylselenocysteine/CPT-11 could be related to increased drug delivery into both tumors (CES1), reduced resistance to SN-38 (ABCC1 and DRG1) in FaDu, and induced Fas ligand apoptosis (TNFSF6) in A253. Irinotecan 84-90 developmentally regulated GTP binding protein 1 Homo sapiens 199-203 15897249-10 2005 In conclusion, the significant increase in the cure rate after methylselenocysteine/CPT-11 could be related to increased drug delivery into both tumors (CES1), reduced resistance to SN-38 (ABCC1 and DRG1) in FaDu, and induced Fas ligand apoptosis (TNFSF6) in A253. Irinotecan 84-90 Fas ligand Homo sapiens 226-236 15897249-10 2005 In conclusion, the significant increase in the cure rate after methylselenocysteine/CPT-11 could be related to increased drug delivery into both tumors (CES1), reduced resistance to SN-38 (ABCC1 and DRG1) in FaDu, and induced Fas ligand apoptosis (TNFSF6) in A253. Irinotecan 84-90 Fas ligand Homo sapiens 248-254 15897249-10 2005 In conclusion, the significant increase in the cure rate after methylselenocysteine/CPT-11 could be related to increased drug delivery into both tumors (CES1), reduced resistance to SN-38 (ABCC1 and DRG1) in FaDu, and induced Fas ligand apoptosis (TNFSF6) in A253. Irinotecan 182-187 ATP binding cassette subfamily C member 1 Homo sapiens 189-194 15897249-7 2005 After methylselenocysteine treatment, the intratumor area under the concentration-time curve of SN-38 increased to a significantly higher level in A253 than in FaDu and was associated with increased expression of CES1 in both tumors. Irinotecan 96-101 carboxylesterase 1 Homo sapiens 213-217 15897249-8 2005 Methylselenocysteine/CPT-11 treatment, compared with CPT-11 alone, resulted in a significant decrease in levels of ABCC1 and DRG1 in FaDu tumors and an increase in levels of CYP3A5 and TNFSF6 in A253 tumors. Irinotecan 21-27 ATP binding cassette subfamily C member 1 Homo sapiens 115-120 15897249-8 2005 Methylselenocysteine/CPT-11 treatment, compared with CPT-11 alone, resulted in a significant decrease in levels of ABCC1 and DRG1 in FaDu tumors and an increase in levels of CYP3A5 and TNFSF6 in A253 tumors. Irinotecan 21-27 developmentally regulated GTP binding protein 1 Homo sapiens 125-129 15897249-8 2005 Methylselenocysteine/CPT-11 treatment, compared with CPT-11 alone, resulted in a significant decrease in levels of ABCC1 and DRG1 in FaDu tumors and an increase in levels of CYP3A5 and TNFSF6 in A253 tumors. Irinotecan 21-27 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 174-180 15897249-8 2005 Methylselenocysteine/CPT-11 treatment, compared with CPT-11 alone, resulted in a significant decrease in levels of ABCC1 and DRG1 in FaDu tumors and an increase in levels of CYP3A5 and TNFSF6 in A253 tumors. Irinotecan 21-27 Fas ligand Homo sapiens 185-191 15996331-0 2005 [Prognostic value of thymidylate synthase, topoisomerase-1 and Ki-67 in advanced colorectal cancer patients on irinotecan and fluorouracil treatment]. Irinotecan 111-121 thymidylate synthetase Homo sapiens 21-41 15846298-7 2005 Combination of HGS-ETR1 with chemotherapeutic agents (topotecan, 5-fluorouracil, and irinotecan) in three independent colon cancer xenograft models resulted in an enhanced antitumour efficacy compared to either agent alone. Irinotecan 85-95 CUGBP Elav-like family member 3 Homo sapiens 19-23 15699047-5 2005 Colony forming assays demonstrated that down-regulation of ATR or Chk1 sensitized cells to SN-38 and camptothecin. Irinotecan 91-96 checkpoint kinase 1 Homo sapiens 66-70 15699047-6 2005 In contrast, ATM and Chk2 had minimal effect of sensitivity to SN-38 or camptothecin. Irinotecan 63-68 checkpoint kinase 2 Homo sapiens 21-25 15699047-7 2005 Additional experiments demonstrated that the Hsp90 inhibitor 17-allylamino-17-demethoxygeldanamycin, which down-regulates Chk1, also sensitized a variety of human carcinoma cell lines to SN-38. Irinotecan 187-192 heat shock protein 90 alpha family class A member 1 Homo sapiens 45-50 15723263-2 2005 The findings reported here demonstrate that SN38 (the active metabolite of CPT-11) induces the tyrosine phosphorylation of EGFR within 5 min, followed by the induction of transcripts and/or proteins of the heparin-binding EGF-like growth factor, amphiregulin, transforming growth factor-alpha, and interlukin-8 (IL-8) in AGS gastric cancer cells. Irinotecan 44-48 epidermal growth factor receptor Homo sapiens 123-127 15801936-3 2005 All patients were genotyped for allelic variants in genes encoding drug metabolizing enzymes (CYP3A4, CYP3A5, UGT1A1) and drug transporters (ABCB1, ABCC2 and ABCG2) that are involved in irinotecan disposition. Irinotecan 186-196 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 110-116 15801936-3 2005 All patients were genotyped for allelic variants in genes encoding drug metabolizing enzymes (CYP3A4, CYP3A5, UGT1A1) and drug transporters (ABCB1, ABCC2 and ABCG2) that are involved in irinotecan disposition. Irinotecan 186-196 ATP binding cassette subfamily B member 1 Homo sapiens 141-146 15801936-3 2005 All patients were genotyped for allelic variants in genes encoding drug metabolizing enzymes (CYP3A4, CYP3A5, UGT1A1) and drug transporters (ABCB1, ABCC2 and ABCG2) that are involved in irinotecan disposition. Irinotecan 186-196 ATP binding cassette subfamily C member 2 Homo sapiens 148-153 15801936-3 2005 All patients were genotyped for allelic variants in genes encoding drug metabolizing enzymes (CYP3A4, CYP3A5, UGT1A1) and drug transporters (ABCB1, ABCC2 and ABCG2) that are involved in irinotecan disposition. Irinotecan 186-196 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 158-163 15723263-2 2005 The findings reported here demonstrate that SN38 (the active metabolite of CPT-11) induces the tyrosine phosphorylation of EGFR within 5 min, followed by the induction of transcripts and/or proteins of the heparin-binding EGF-like growth factor, amphiregulin, transforming growth factor-alpha, and interlukin-8 (IL-8) in AGS gastric cancer cells. Irinotecan 75-81 epidermal growth factor receptor Homo sapiens 123-127 15801936-6 2005 The C(max) of irinotecan was significantly lower in patients carrying the CC genotype at exon 26 of the ABCB1 gene compared with those harbouring at least one variant allele (P = 0.047). Irinotecan 14-24 ATP binding cassette subfamily B member 1 Homo sapiens 104-109 15801936-7 2005 Patients harbouring the wild type ABCG2 CTCA genotype were associated with significantly higher values for relative extent of conversion (REC) of irinotecan to SN-38 compared with patients carrying at least one deletion CTCA allele (P = 0.019). Irinotecan 146-156 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 34-39 15855893-5 2005 Chimeric monoclonal antibody therapy directed at the epidermal growth factor receptor was associated with radiographic responses in a significant minority of patients with irinotecan-refractory colon cancer in a randomized phase II study of patients with irinotecan-refractory disease. Irinotecan 172-182 epidermal growth factor receptor Homo sapiens 53-85 15801936-7 2005 Patients harbouring the wild type ABCG2 CTCA genotype were associated with significantly higher values for relative extent of conversion (REC) of irinotecan to SN-38 compared with patients carrying at least one deletion CTCA allele (P = 0.019). Irinotecan 160-165 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 34-39 15801936-8 2005 CONCLUSIONS: The present exploratory study shows that genetic polymorphisms in drug transporter genes, particularly in ABCB1 and ABCG2 genes, may be important in influencing the pharmacokinetics of irinotecan and its metabolites. Irinotecan 198-208 ATP binding cassette subfamily B member 1 Homo sapiens 119-124 15801936-8 2005 CONCLUSIONS: The present exploratory study shows that genetic polymorphisms in drug transporter genes, particularly in ABCB1 and ABCG2 genes, may be important in influencing the pharmacokinetics of irinotecan and its metabolites. Irinotecan 198-208 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 129-134 15801936-9 2005 The predictive value of the identified allelic variants in the ABCG2 and ABCB1 genes on irinotecan disposition should be further investigated in a larger patient population as well as in other ethnic populations. Irinotecan 88-98 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 63-68 15801936-9 2005 The predictive value of the identified allelic variants in the ABCG2 and ABCB1 genes on irinotecan disposition should be further investigated in a larger patient population as well as in other ethnic populations. Irinotecan 88-98 ATP binding cassette subfamily B member 1 Homo sapiens 73-78 15855893-5 2005 Chimeric monoclonal antibody therapy directed at the epidermal growth factor receptor was associated with radiographic responses in a significant minority of patients with irinotecan-refractory colon cancer in a randomized phase II study of patients with irinotecan-refractory disease. Irinotecan 255-265 epidermal growth factor receptor Homo sapiens 53-85 15583935-0 2005 Anti-angiogenic effects of SN38 (active metabolite of irinotecan): inhibition of hypoxia-inducible factor 1 alpha (HIF-1alpha)/vascular endothelial growth factor (VEGF) expression of glioma and growth of endothelial cells. Irinotecan 27-31 hypoxia inducible factor 1 subunit alpha Homo sapiens 81-113 15827327-0 2005 Expression of multidrug transporter MRP4/ABCC4 is a marker of poor prognosis in neuroblastoma and confers resistance to irinotecan in vitro. Irinotecan 120-130 ATP binding cassette subfamily C member 4 Homo sapiens 36-40 15827327-0 2005 Expression of multidrug transporter MRP4/ABCC4 is a marker of poor prognosis in neuroblastoma and confers resistance to irinotecan in vitro. Irinotecan 120-130 ATP binding cassette subfamily C member 4 Homo sapiens 41-46 15583935-0 2005 Anti-angiogenic effects of SN38 (active metabolite of irinotecan): inhibition of hypoxia-inducible factor 1 alpha (HIF-1alpha)/vascular endothelial growth factor (VEGF) expression of glioma and growth of endothelial cells. Irinotecan 27-31 hypoxia inducible factor 1 subunit alpha Homo sapiens 115-125 15583935-0 2005 Anti-angiogenic effects of SN38 (active metabolite of irinotecan): inhibition of hypoxia-inducible factor 1 alpha (HIF-1alpha)/vascular endothelial growth factor (VEGF) expression of glioma and growth of endothelial cells. Irinotecan 27-31 vascular endothelial growth factor A Homo sapiens 127-161 15583935-0 2005 Anti-angiogenic effects of SN38 (active metabolite of irinotecan): inhibition of hypoxia-inducible factor 1 alpha (HIF-1alpha)/vascular endothelial growth factor (VEGF) expression of glioma and growth of endothelial cells. Irinotecan 27-31 vascular endothelial growth factor A Homo sapiens 163-167 15583935-0 2005 Anti-angiogenic effects of SN38 (active metabolite of irinotecan): inhibition of hypoxia-inducible factor 1 alpha (HIF-1alpha)/vascular endothelial growth factor (VEGF) expression of glioma and growth of endothelial cells. Irinotecan 54-64 hypoxia inducible factor 1 subunit alpha Homo sapiens 81-113 15583935-0 2005 Anti-angiogenic effects of SN38 (active metabolite of irinotecan): inhibition of hypoxia-inducible factor 1 alpha (HIF-1alpha)/vascular endothelial growth factor (VEGF) expression of glioma and growth of endothelial cells. Irinotecan 54-64 hypoxia inducible factor 1 subunit alpha Homo sapiens 115-125 15583935-0 2005 Anti-angiogenic effects of SN38 (active metabolite of irinotecan): inhibition of hypoxia-inducible factor 1 alpha (HIF-1alpha)/vascular endothelial growth factor (VEGF) expression of glioma and growth of endothelial cells. Irinotecan 54-64 vascular endothelial growth factor A Homo sapiens 127-161 15583935-0 2005 Anti-angiogenic effects of SN38 (active metabolite of irinotecan): inhibition of hypoxia-inducible factor 1 alpha (HIF-1alpha)/vascular endothelial growth factor (VEGF) expression of glioma and growth of endothelial cells. Irinotecan 54-64 vascular endothelial growth factor A Homo sapiens 163-167 15788689-7 2005 The JNK inhibitor SP600125 substantially decreased the activation of caspases and apoptosis induced by MSeA combination with SN38 or etoposide and completely blocked these events induced by MSeA/paclitaxel. Irinotecan 125-129 mitogen-activated protein kinase 8 Homo sapiens 4-7 15788689-7 2005 The JNK inhibitor SP600125 substantially decreased the activation of caspases and apoptosis induced by MSeA combination with SN38 or etoposide and completely blocked these events induced by MSeA/paclitaxel. Irinotecan 125-129 caspase 8 Homo sapiens 69-77 15788689-10 2005 A JNK-independent suppression of survivin by SN38 and etoposide, but not by paclitaxel, was also observed. Irinotecan 45-49 mitogen-activated protein kinase 8 Homo sapiens 2-5 15608127-7 2005 Among the variants examined, OATP1B1*15 (N130D and V174A; reported allele frequency 10-15%) exhibited decreased transport activities for SN-38 as well as pravastatin, estrone-3-sulfate, and estradiol-17beta-glucuronide. Irinotecan 137-142 solute carrier organic anion transporter family member 1B1 Homo sapiens 29-36 15572581-10 2005 The intrinsic clearance of 7-ethyl-10-hydroxycamptothecin (SN-38) by P229L UGT1A1 expressed in COS-1 cells was about 3% of the wild type. Irinotecan 27-57 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 75-81 15572581-10 2005 The intrinsic clearance of 7-ethyl-10-hydroxycamptothecin (SN-38) by P229L UGT1A1 expressed in COS-1 cells was about 3% of the wild type. Irinotecan 59-64 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 75-81 15608127-0 2005 Role of organic anion transporter OATP1B1 (OATP-C) in hepatic uptake of irinotecan and its active metabolite, 7-ethyl-10-hydroxycamptothecin: in vitro evidence and effect of single nucleotide polymorphisms. Irinotecan 72-82 solute carrier organic anion transporter family member 1B1 Homo sapiens 34-41 15608127-0 2005 Role of organic anion transporter OATP1B1 (OATP-C) in hepatic uptake of irinotecan and its active metabolite, 7-ethyl-10-hydroxycamptothecin: in vitro evidence and effect of single nucleotide polymorphisms. Irinotecan 72-82 solute carrier organic anion transporter family member 1B1 Homo sapiens 43-49 15608127-8 2005 This study is the first to yield evidence that OATP1B1 is involved in the hepatic disposition of SN-38 and that genetic polymorphisms of OATP1B1 may contribute to the known interpatient variability in disposition of irinotecan. Irinotecan 97-102 solute carrier organic anion transporter family member 1B1 Homo sapiens 47-54 15608127-0 2005 Role of organic anion transporter OATP1B1 (OATP-C) in hepatic uptake of irinotecan and its active metabolite, 7-ethyl-10-hydroxycamptothecin: in vitro evidence and effect of single nucleotide polymorphisms. Irinotecan 110-140 solute carrier organic anion transporter family member 1B1 Homo sapiens 34-41 15608127-0 2005 Role of organic anion transporter OATP1B1 (OATP-C) in hepatic uptake of irinotecan and its active metabolite, 7-ethyl-10-hydroxycamptothecin: in vitro evidence and effect of single nucleotide polymorphisms. Irinotecan 110-140 solute carrier organic anion transporter family member 1B1 Homo sapiens 43-49 15608127-8 2005 This study is the first to yield evidence that OATP1B1 is involved in the hepatic disposition of SN-38 and that genetic polymorphisms of OATP1B1 may contribute to the known interpatient variability in disposition of irinotecan. Irinotecan 216-226 solute carrier organic anion transporter family member 1B1 Homo sapiens 137-144 15608127-3 2005 Here, we examined the contribution of an organic anion-transporting polypeptide, OATP1B1 (OATP-C), which transports a variety of drugs and their metabolites from blood to liver in humans, to the hepatic disposition of irinotecan, SN-38, and its glucuronide conjugate (SN-38G) by using HEK293 cells stably transfected with SLCO1B1*1a (OATP-C*1a) coding wild-type OATP1B1. Irinotecan 218-228 solute carrier organic anion transporter family member 1B1 Homo sapiens 81-88 15608127-3 2005 Here, we examined the contribution of an organic anion-transporting polypeptide, OATP1B1 (OATP-C), which transports a variety of drugs and their metabolites from blood to liver in humans, to the hepatic disposition of irinotecan, SN-38, and its glucuronide conjugate (SN-38G) by using HEK293 cells stably transfected with SLCO1B1*1a (OATP-C*1a) coding wild-type OATP1B1. Irinotecan 218-228 solute carrier organic anion transporter family member 1B1 Homo sapiens 90-96 15608127-3 2005 Here, we examined the contribution of an organic anion-transporting polypeptide, OATP1B1 (OATP-C), which transports a variety of drugs and their metabolites from blood to liver in humans, to the hepatic disposition of irinotecan, SN-38, and its glucuronide conjugate (SN-38G) by using HEK293 cells stably transfected with SLCO1B1*1a (OATP-C*1a) coding wild-type OATP1B1. Irinotecan 230-235 solute carrier organic anion transporter family member 1B1 Homo sapiens 81-88 15608127-3 2005 Here, we examined the contribution of an organic anion-transporting polypeptide, OATP1B1 (OATP-C), which transports a variety of drugs and their metabolites from blood to liver in humans, to the hepatic disposition of irinotecan, SN-38, and its glucuronide conjugate (SN-38G) by using HEK293 cells stably transfected with SLCO1B1*1a (OATP-C*1a) coding wild-type OATP1B1. Irinotecan 230-235 solute carrier organic anion transporter family member 1B1 Homo sapiens 90-96 15608127-3 2005 Here, we examined the contribution of an organic anion-transporting polypeptide, OATP1B1 (OATP-C), which transports a variety of drugs and their metabolites from blood to liver in humans, to the hepatic disposition of irinotecan, SN-38, and its glucuronide conjugate (SN-38G) by using HEK293 cells stably transfected with SLCO1B1*1a (OATP-C*1a) coding wild-type OATP1B1. Irinotecan 268-273 solute carrier organic anion transporter family member 1B1 Homo sapiens 81-88 15608127-3 2005 Here, we examined the contribution of an organic anion-transporting polypeptide, OATP1B1 (OATP-C), which transports a variety of drugs and their metabolites from blood to liver in humans, to the hepatic disposition of irinotecan, SN-38, and its glucuronide conjugate (SN-38G) by using HEK293 cells stably transfected with SLCO1B1*1a (OATP-C*1a) coding wild-type OATP1B1. Irinotecan 268-273 solute carrier organic anion transporter family member 1B1 Homo sapiens 90-96 15608127-6 2005 Moreover, SN-38 exhibited a significant inhibitory effect on OATP1B1-mediated uptake of [(3)H]estrone-3-sulfate. Irinotecan 10-15 solute carrier organic anion transporter family member 1B1 Homo sapiens 61-68 15668287-9 2005 CONCLUSIONS: The administration of irinotecan with docetaxel in platinum-refractory NSCLC prolonged TTP, but did not improve significantly RR, median survival or 1-year survival. Irinotecan 35-45 ZFP36 ring finger protein Homo sapiens 100-103 15686910-3 2005 Furthermore bisbenzyl 4 potentiated the cytotoxicity of SN-38 in BCRP-transduced K562 (K562/BCRP) cells. Irinotecan 56-61 BCR pseudogene 1 Homo sapiens 65-69 15686910-3 2005 Furthermore bisbenzyl 4 potentiated the cytotoxicity of SN-38 in BCRP-transduced K562 (K562/BCRP) cells. Irinotecan 56-61 BCR pseudogene 1 Homo sapiens 92-96 15709193-0 2005 UGT1A7 and UGT1A9 polymorphisms predict response and toxicity in colorectal cancer patients treated with capecitabine/irinotecan. Irinotecan 118-128 UDP glucuronosyltransferase family 1 member A7 Homo sapiens 0-6 15709193-0 2005 UGT1A7 and UGT1A9 polymorphisms predict response and toxicity in colorectal cancer patients treated with capecitabine/irinotecan. Irinotecan 118-128 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 11-17 15709193-2 2005 We hypothesized that germline polymorphisms within genes related to drug target (thymidylate synthase) or metabolizing enzymes (UDP-glucuronosyltransferase, UGT) would impact response and toxicity to the combination of capecitabine plus irinotecan (CPT-11). Irinotecan 237-247 thymidylate synthetase Homo sapiens 81-101 15709193-2 2005 We hypothesized that germline polymorphisms within genes related to drug target (thymidylate synthase) or metabolizing enzymes (UDP-glucuronosyltransferase, UGT) would impact response and toxicity to the combination of capecitabine plus irinotecan (CPT-11). Irinotecan 237-247 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 157-160 15709193-2 2005 We hypothesized that germline polymorphisms within genes related to drug target (thymidylate synthase) or metabolizing enzymes (UDP-glucuronosyltransferase, UGT) would impact response and toxicity to the combination of capecitabine plus irinotecan (CPT-11). Irinotecan 249-255 thymidylate synthetase Homo sapiens 81-101 15709193-2 2005 We hypothesized that germline polymorphisms within genes related to drug target (thymidylate synthase) or metabolizing enzymes (UDP-glucuronosyltransferase, UGT) would impact response and toxicity to the combination of capecitabine plus irinotecan (CPT-11). Irinotecan 249-255 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 157-160 15709193-10 2005 CONCLUSIONS: These data strongly suggest that UGT1A7 and/or UGT1A9 genotypes may be predictors of response and toxicity in CRC patients treated with capecitabine plus irinotecan. Irinotecan 167-177 UDP glucuronosyltransferase family 1 member A7 Homo sapiens 46-52 15709193-10 2005 CONCLUSIONS: These data strongly suggest that UGT1A7 and/or UGT1A9 genotypes may be predictors of response and toxicity in CRC patients treated with capecitabine plus irinotecan. Irinotecan 167-177 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 60-66 15757419-4 2005 S-1-based combination therapies with other promising drugs, including cis-platin, irinotecan and taxanes, are expected to yield good results. Irinotecan 82-92 proteasome 26S subunit, non-ATPase 1 Homo sapiens 0-3 15821783-23 2005 Results of a large phase II study have shown response when used in combination with irinotecan in 22.9% of patients with EGFR-expressing, irinotecan-refractory, colorectal cancer. Irinotecan 84-94 epidermal growth factor receptor Homo sapiens 121-125 15821783-23 2005 Results of a large phase II study have shown response when used in combination with irinotecan in 22.9% of patients with EGFR-expressing, irinotecan-refractory, colorectal cancer. Irinotecan 138-148 epidermal growth factor receptor Homo sapiens 121-125 15558802-1 2005 BACKGROUND: In the current study, the authors report a Phase II trial of irinotecan (CPT-11), a topoisomerase I inhibitor active against malignant glioma (MG), with celecoxib, a selective COX-2 inhibitor, among MG patients with recurrent disease. Irinotecan 73-83 mitochondrially encoded cytochrome c oxidase II Homo sapiens 188-193 15709211-14 2005 There it was reported that the combination of 5-fluorouracil, leucovorin, and irinotecan (Camptosar; Pfizer Pharmaceuticals; New York, NY) with anti-vascular endothelial growth factor antibody (bevacizumab-Avastin; Genentech, Inc.; South San Francisco, CA) was superior to the chemotherapy regimen alone when used to treat patients with metastatic colorectal cancer. Irinotecan 78-88 vascular endothelial growth factor A Homo sapiens 149-183 15709211-14 2005 There it was reported that the combination of 5-fluorouracil, leucovorin, and irinotecan (Camptosar; Pfizer Pharmaceuticals; New York, NY) with anti-vascular endothelial growth factor antibody (bevacizumab-Avastin; Genentech, Inc.; South San Francisco, CA) was superior to the chemotherapy regimen alone when used to treat patients with metastatic colorectal cancer. Irinotecan 90-99 vascular endothelial growth factor A Homo sapiens 149-183 15709212-4 2005 There exist several clinically relevant examples of the utility of pharmacogenomics that associate specific genetic polymorphisms in drug metabolizing enzymes (e.g., TPMT, UGT1A1, DPD), drug transporters (MDR1), and drug target enzymes (TS) with clinical outcomes in patients treated with commonly prescribed chemotherapy drugs, such as 5-fluorouracil and irinotecan (Camptosar; Pfizer Pharmaceuticals; New York, NY http://www.pfizer.com). Irinotecan 356-366 thiopurine S-methyltransferase Homo sapiens 166-170 15709212-4 2005 There exist several clinically relevant examples of the utility of pharmacogenomics that associate specific genetic polymorphisms in drug metabolizing enzymes (e.g., TPMT, UGT1A1, DPD), drug transporters (MDR1), and drug target enzymes (TS) with clinical outcomes in patients treated with commonly prescribed chemotherapy drugs, such as 5-fluorouracil and irinotecan (Camptosar; Pfizer Pharmaceuticals; New York, NY http://www.pfizer.com). Irinotecan 368-377 thiopurine S-methyltransferase Homo sapiens 166-170 15695404-1 2005 Breast cancer resistance protein (BCRP)/ABCG2 mediates concurrent resistance to chemotherapeutic agents, such as 7-ethyl-10-hydroxycamptothecin (SN-38), mitoxantrone, and topotecan, by pumping them out of cells. Irinotecan 113-143 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 0-32 15695404-1 2005 Breast cancer resistance protein (BCRP)/ABCG2 mediates concurrent resistance to chemotherapeutic agents, such as 7-ethyl-10-hydroxycamptothecin (SN-38), mitoxantrone, and topotecan, by pumping them out of cells. Irinotecan 113-143 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 34-38 15695404-1 2005 Breast cancer resistance protein (BCRP)/ABCG2 mediates concurrent resistance to chemotherapeutic agents, such as 7-ethyl-10-hydroxycamptothecin (SN-38), mitoxantrone, and topotecan, by pumping them out of cells. Irinotecan 113-143 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 40-45 15695404-1 2005 Breast cancer resistance protein (BCRP)/ABCG2 mediates concurrent resistance to chemotherapeutic agents, such as 7-ethyl-10-hydroxycamptothecin (SN-38), mitoxantrone, and topotecan, by pumping them out of cells. Irinotecan 145-150 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 0-32 15695404-1 2005 Breast cancer resistance protein (BCRP)/ABCG2 mediates concurrent resistance to chemotherapeutic agents, such as 7-ethyl-10-hydroxycamptothecin (SN-38), mitoxantrone, and topotecan, by pumping them out of cells. Irinotecan 145-150 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 34-38 15695404-1 2005 Breast cancer resistance protein (BCRP)/ABCG2 mediates concurrent resistance to chemotherapeutic agents, such as 7-ethyl-10-hydroxycamptothecin (SN-38), mitoxantrone, and topotecan, by pumping them out of cells. Irinotecan 145-150 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 40-45 15695404-5 2005 E(2) potentiates the cytotoxicity of SN-38, but not vincristine, in MCF-7/BCRP cells significantly, and increases cellular topotecan uptake in MCF-7 and MCF-7/BCRP cells. Irinotecan 37-42 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 74-78 15727486-10 2005 The enzymes responsible for the activation, metabolism and mechanism of action of irinotecan, namely carboxylesterase 2, cytochrome P450 (CYP) 3A4, uridine diphosphate glucuronosyltransferase isoform 1A1 (UGT1A1), and topoisomerase-I, also exhibit variable interindividual activity. Irinotecan 82-92 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 121-146 15727486-10 2005 The enzymes responsible for the activation, metabolism and mechanism of action of irinotecan, namely carboxylesterase 2, cytochrome P450 (CYP) 3A4, uridine diphosphate glucuronosyltransferase isoform 1A1 (UGT1A1), and topoisomerase-I, also exhibit variable interindividual activity. Irinotecan 82-92 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 148-203 15727486-10 2005 The enzymes responsible for the activation, metabolism and mechanism of action of irinotecan, namely carboxylesterase 2, cytochrome P450 (CYP) 3A4, uridine diphosphate glucuronosyltransferase isoform 1A1 (UGT1A1), and topoisomerase-I, also exhibit variable interindividual activity. Irinotecan 82-92 carboxylesterase 2 Homo sapiens 101-119 15727486-10 2005 The enzymes responsible for the activation, metabolism and mechanism of action of irinotecan, namely carboxylesterase 2, cytochrome P450 (CYP) 3A4, uridine diphosphate glucuronosyltransferase isoform 1A1 (UGT1A1), and topoisomerase-I, also exhibit variable interindividual activity. Irinotecan 82-92 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 205-211 15727486-13 2005 CYP3A4 and UGT1A1 activity levels might be predictive of irinotecan toxicity rather than efficacy. Irinotecan 57-67 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 0-6 15727486-13 2005 CYP3A4 and UGT1A1 activity levels might be predictive of irinotecan toxicity rather than efficacy. Irinotecan 57-67 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 11-17 15813675-2 2005 Cancer cells overexpressing the ABCG2 gene show multidrug resistance to mitoxantrone-, methotrexate-, doxorubicin-, and camptothecin-based anticancer drugs, such as topotecan and SN-38. Irinotecan 179-184 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 32-37 15720263-8 2005 Uridine diphosphate glucuronosyltransferase isoform 1A1 (UGT1A1) genotype has been reported to be associated with time to progression and survival in patients treated with irinotecan. Irinotecan 172-182 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-55 15613901-11 2005 This retrospective study showed a high resectability rate, and a prolonged TTP and OS in patients with ACRC after induction treatment with irinotecan-based chemotherapy. Irinotecan 139-149 ZFP36 ring finger protein Homo sapiens 75-78 15720263-8 2005 Uridine diphosphate glucuronosyltransferase isoform 1A1 (UGT1A1) genotype has been reported to be associated with time to progression and survival in patients treated with irinotecan. Irinotecan 172-182 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 57-63 16280599-6 2005 Moreover, both resveratrol and CPT-11, which are known to arrest cell-cycle progression, elevated egr-1 mRNA levels in glioma cells. Irinotecan 31-37 early growth response 1 Rattus norvegicus 98-103 16257834-2 2005 Irinotecan undergoes drug metabolism to form an active SN-38, which is further converted to its beta-glucuronide by UDP-glucuronosyltransferase (UGT) 1A1. Irinotecan 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 116-153 16257834-2 2005 Irinotecan undergoes drug metabolism to form an active SN-38, which is further converted to its beta-glucuronide by UDP-glucuronosyltransferase (UGT) 1A1. Irinotecan 55-60 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 116-153 16257834-3 2005 A variant in the promoter of UGT1A1 gene, UGT1A1*28 allele, has been extensively studied, and pharmacogenetic relationships between the variant and ADR to irinotecan have been reported. Irinotecan 155-165 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 29-35 16257834-3 2005 A variant in the promoter of UGT1A1 gene, UGT1A1*28 allele, has been extensively studied, and pharmacogenetic relationships between the variant and ADR to irinotecan have been reported. Irinotecan 155-165 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 42-48 16257834-4 2005 A case-control study of Japanese cancer patients demonstrated that the patients having UGT1A1*28 were at significantly increased risk of severe ADR to irinotecan. Irinotecan 151-161 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 87-93 16257834-5 2005 To date, genetic variations of the UGT1A1 gene is the most important hereditary factor to predict severe ADR to irinotecan. Irinotecan 112-122 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 35-41 16257834-7 2005 At present, irinotecan chemotherapy based on a patient"s UGT1A1 genetic status is scientifically reasonable. Irinotecan 12-22 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 57-63 16399345-2 2005 In addition to bilirubin, UGT1A1 conjugates various endogenous and exogenous lipophilic compounds such as estrogens and the active metabolite of the anticancer drug irinotecan SN-38. Irinotecan 165-175 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 26-32 16399345-2 2005 In addition to bilirubin, UGT1A1 conjugates various endogenous and exogenous lipophilic compounds such as estrogens and the active metabolite of the anticancer drug irinotecan SN-38. Irinotecan 176-181 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 26-32 16399345-3 2005 Thus, activation by specific inducers of the UGT1A1 gene is critical in treating patients with unconjugated hyperbili-rubinemia and in preventing side effects of drug treatment such as SN-38-induced toxicity. Irinotecan 185-190 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 45-51 16491956-0 2005 Irinotecan/5-fluorouracil combination induces alterations in mitochondrial membrane potential and caspases on colon cancer cell lines. Irinotecan 0-10 caspase 8 Homo sapiens 98-106 16491956-5 2005 In this study, we verified whether the collapse in mitochondrial membrane and the activation of caspases is responsible for increased apoptosis observed with CPT-11/5-FU treatment. Irinotecan 158-164 caspase 8 Homo sapiens 96-104 16491956-8 2005 In these cells, both caspase-3 and -9 were involved in the activation of apoptosis after CPT-11/5-FU treatment. Irinotecan 89-95 caspase 3 Homo sapiens 21-37 16491956-12 2005 In spite of the differences among the cell lines, these results indicated that the increase in apoptosis in HT-29 cells observed with CPT-11 followed by 5-FU treatment could be explained by a disruption in mitochondria membrane potential that induced caspases activation. Irinotecan 134-140 caspase 8 Homo sapiens 251-259 15604280-0 2004 Elimination of hepatic metastases of colon cancer cells via p53-independent cross-talk between irinotecan and Apo2 ligand/TRAIL. Irinotecan 95-105 tumor protein p53 Homo sapiens 60-63 16020968-13 2005 CONCLUSIONS: Biweekly DTX/CPT-11 with G-CSF support is a well-tolerated and highly effective approach in anthracycline-/paclitaxel-pretreated patients. Irinotecan 26-32 colony stimulating factor 3 Homo sapiens 38-43 15864124-0 2005 Genetic polymorphism in the phenobarbital-responsive enhancer module of the UDP-glucuronosyltransferase 1A1 gene and irinotecan toxicity. Irinotecan 117-127 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 76-107 15864124-1 2005 Genetic polymorphism of the UDP-glucuronosyltransferase (UGT) 1A1 gene is associated with the decreased glucuronidation activity of an active metabolite of irinotecan, SN-38, and UGT1A1*28 has been shown as a predictive factor for irinotecan toxicity. Irinotecan 156-166 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 28-65 15864124-1 2005 Genetic polymorphism of the UDP-glucuronosyltransferase (UGT) 1A1 gene is associated with the decreased glucuronidation activity of an active metabolite of irinotecan, SN-38, and UGT1A1*28 has been shown as a predictive factor for irinotecan toxicity. Irinotecan 156-166 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 179-185 15864124-1 2005 Genetic polymorphism of the UDP-glucuronosyltransferase (UGT) 1A1 gene is associated with the decreased glucuronidation activity of an active metabolite of irinotecan, SN-38, and UGT1A1*28 has been shown as a predictive factor for irinotecan toxicity. Irinotecan 168-173 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 28-65 15864124-1 2005 Genetic polymorphism of the UDP-glucuronosyltransferase (UGT) 1A1 gene is associated with the decreased glucuronidation activity of an active metabolite of irinotecan, SN-38, and UGT1A1*28 has been shown as a predictive factor for irinotecan toxicity. Irinotecan 231-241 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 28-65 15864124-1 2005 Genetic polymorphism of the UDP-glucuronosyltransferase (UGT) 1A1 gene is associated with the decreased glucuronidation activity of an active metabolite of irinotecan, SN-38, and UGT1A1*28 has been shown as a predictive factor for irinotecan toxicity. Irinotecan 231-241 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 179-185 15864124-12 2005 The determination of T-3279G and UGT1A1*28 genotypes might be clinically useful in predicting severe irinotecan toxicity in cancer patients. Irinotecan 101-111 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-39 15604285-5 2004 Surprisingly, the RNase L-deficient cells were also highly resistant to apoptosis by combination treatments with a topoisomerase (Topo) I inhibitor (camptothecin, topotecan, or SN-38) and tumor necrosis factor-related apoptosis-inducing ligand [TRAIL (Apo2L)]. Irinotecan 177-182 ribonuclease L Homo sapiens 18-25 15604280-3 2004 Whereas irinotecan-induced up-regulation of the proapoptotic proteins PUMA and Noxa requires p53, we find that irinotecan inhibits Janus kinase 2 (JAK2)-signal transducer and activator of transcription 3 and 5 (STAT3/5) signaling in both p53-proficient and p53-deficient tumor cells. Irinotecan 111-121 tumor protein p53 Homo sapiens 238-241 15604280-4 2004 We show that irinotecan inhibits JAK2-STAT3/5-dependent expression of survival proteins (Bcl-x(L) and XIAP) and cooperates with Apo2 ligand/tumor necrosis factor-related apoptosis-inducing ligand (Apo2L/TRAIL) to facilitate p53-independent apoptosis of colon cancer cells. Irinotecan 13-23 Janus kinase 2 Homo sapiens 33-37 15604280-4 2004 We show that irinotecan inhibits JAK2-STAT3/5-dependent expression of survival proteins (Bcl-x(L) and XIAP) and cooperates with Apo2 ligand/tumor necrosis factor-related apoptosis-inducing ligand (Apo2L/TRAIL) to facilitate p53-independent apoptosis of colon cancer cells. Irinotecan 13-23 signal transducer and activator of transcription 3 Homo sapiens 38-45 15604280-2 2004 Using isogenic human colon cancer cells that differ only in their p53 status, we demonstrate that loss of p53 renders tumor cells relatively resistant to the topoisomerase I inhibitor, irinotecan. Irinotecan 185-195 tumor protein p53 Homo sapiens 106-109 15604280-4 2004 We show that irinotecan inhibits JAK2-STAT3/5-dependent expression of survival proteins (Bcl-x(L) and XIAP) and cooperates with Apo2 ligand/tumor necrosis factor-related apoptosis-inducing ligand (Apo2L/TRAIL) to facilitate p53-independent apoptosis of colon cancer cells. Irinotecan 13-23 BCL2 like 1 Homo sapiens 89-97 15604280-3 2004 Whereas irinotecan-induced up-regulation of the proapoptotic proteins PUMA and Noxa requires p53, we find that irinotecan inhibits Janus kinase 2 (JAK2)-signal transducer and activator of transcription 3 and 5 (STAT3/5) signaling in both p53-proficient and p53-deficient tumor cells. Irinotecan 8-18 tumor protein p53 Homo sapiens 93-96 15604280-4 2004 We show that irinotecan inhibits JAK2-STAT3/5-dependent expression of survival proteins (Bcl-x(L) and XIAP) and cooperates with Apo2 ligand/tumor necrosis factor-related apoptosis-inducing ligand (Apo2L/TRAIL) to facilitate p53-independent apoptosis of colon cancer cells. Irinotecan 13-23 X-linked inhibitor of apoptosis Homo sapiens 102-106 15604280-4 2004 We show that irinotecan inhibits JAK2-STAT3/5-dependent expression of survival proteins (Bcl-x(L) and XIAP) and cooperates with Apo2 ligand/tumor necrosis factor-related apoptosis-inducing ligand (Apo2L/TRAIL) to facilitate p53-independent apoptosis of colon cancer cells. Irinotecan 13-23 TNF receptor superfamily member 10a Homo sapiens 128-132 15604280-3 2004 Whereas irinotecan-induced up-regulation of the proapoptotic proteins PUMA and Noxa requires p53, we find that irinotecan inhibits Janus kinase 2 (JAK2)-signal transducer and activator of transcription 3 and 5 (STAT3/5) signaling in both p53-proficient and p53-deficient tumor cells. Irinotecan 8-18 Janus kinase 2 Homo sapiens 147-151 15604280-4 2004 We show that irinotecan inhibits JAK2-STAT3/5-dependent expression of survival proteins (Bcl-x(L) and XIAP) and cooperates with Apo2 ligand/tumor necrosis factor-related apoptosis-inducing ligand (Apo2L/TRAIL) to facilitate p53-independent apoptosis of colon cancer cells. Irinotecan 13-23 TNF superfamily member 10 Homo sapiens 197-202 15604280-4 2004 We show that irinotecan inhibits JAK2-STAT3/5-dependent expression of survival proteins (Bcl-x(L) and XIAP) and cooperates with Apo2 ligand/tumor necrosis factor-related apoptosis-inducing ligand (Apo2L/TRAIL) to facilitate p53-independent apoptosis of colon cancer cells. Irinotecan 13-23 TNF superfamily member 10 Homo sapiens 203-208 15604280-4 2004 We show that irinotecan inhibits JAK2-STAT3/5-dependent expression of survival proteins (Bcl-x(L) and XIAP) and cooperates with Apo2 ligand/tumor necrosis factor-related apoptosis-inducing ligand (Apo2L/TRAIL) to facilitate p53-independent apoptosis of colon cancer cells. Irinotecan 13-23 tumor protein p53 Homo sapiens 224-227 15604280-5 2004 Whereas xenografts of p53-deficient colon cancer cells are relatively resistant to irinotecan compared with their p53-proficient counterparts, combined treatment with irinotecan and Apo2L/TRAIL eliminates hepatic metastases of both p53-proficient and p53-deficient cancer cells in vivo and significantly improves the survival of animals relative to treatment with either agent alone. Irinotecan 83-93 tumor protein p53 Homo sapiens 22-25 15604280-5 2004 Whereas xenografts of p53-deficient colon cancer cells are relatively resistant to irinotecan compared with their p53-proficient counterparts, combined treatment with irinotecan and Apo2L/TRAIL eliminates hepatic metastases of both p53-proficient and p53-deficient cancer cells in vivo and significantly improves the survival of animals relative to treatment with either agent alone. Irinotecan 83-93 TNF superfamily member 10 Homo sapiens 188-193 15604280-5 2004 Whereas xenografts of p53-deficient colon cancer cells are relatively resistant to irinotecan compared with their p53-proficient counterparts, combined treatment with irinotecan and Apo2L/TRAIL eliminates hepatic metastases of both p53-proficient and p53-deficient cancer cells in vivo and significantly improves the survival of animals relative to treatment with either agent alone. Irinotecan 167-177 tumor protein p53 Homo sapiens 114-117 15604280-6 2004 Although the synergy between chemotherapy and Apo2L/TRAIL has been ascribed to p53, our data demonstrate that irinotecan enhances Apo2L/TRAIL-induced apoptosis of tumor cells via a distinct p53-independent mechanism involving inhibition of JAK2-STAT3/5 signaling. Irinotecan 110-120 TNF superfamily member 10 Homo sapiens 130-135 15604280-6 2004 Although the synergy between chemotherapy and Apo2L/TRAIL has been ascribed to p53, our data demonstrate that irinotecan enhances Apo2L/TRAIL-induced apoptosis of tumor cells via a distinct p53-independent mechanism involving inhibition of JAK2-STAT3/5 signaling. Irinotecan 110-120 TNF superfamily member 10 Homo sapiens 136-141 15604280-6 2004 Although the synergy between chemotherapy and Apo2L/TRAIL has been ascribed to p53, our data demonstrate that irinotecan enhances Apo2L/TRAIL-induced apoptosis of tumor cells via a distinct p53-independent mechanism involving inhibition of JAK2-STAT3/5 signaling. Irinotecan 110-120 tumor protein p53 Homo sapiens 190-193 15604280-6 2004 Although the synergy between chemotherapy and Apo2L/TRAIL has been ascribed to p53, our data demonstrate that irinotecan enhances Apo2L/TRAIL-induced apoptosis of tumor cells via a distinct p53-independent mechanism involving inhibition of JAK2-STAT3/5 signaling. Irinotecan 110-120 Janus kinase 2 Homo sapiens 240-244 15604280-6 2004 Although the synergy between chemotherapy and Apo2L/TRAIL has been ascribed to p53, our data demonstrate that irinotecan enhances Apo2L/TRAIL-induced apoptosis of tumor cells via a distinct p53-independent mechanism involving inhibition of JAK2-STAT3/5 signaling. Irinotecan 110-120 signal transducer and activator of transcription 3 Homo sapiens 245-252 15604280-3 2004 Whereas irinotecan-induced up-regulation of the proapoptotic proteins PUMA and Noxa requires p53, we find that irinotecan inhibits Janus kinase 2 (JAK2)-signal transducer and activator of transcription 3 and 5 (STAT3/5) signaling in both p53-proficient and p53-deficient tumor cells. Irinotecan 8-18 signal transducer and activator of transcription 3 Homo sapiens 211-218 15604280-7 2004 These findings identify a novel p53-independent channel of cross-talk between topoisomerase I inhibitors and Apo2L/TRAIL and suggest that the addition of Apo2L/TRAIL can improve the therapeutic index of irinotecan against both p53-proficient and p53-deficient colorectal cancers, including those that have metastasized to the liver. Irinotecan 203-213 tumor protein p53 Homo sapiens 32-35 15604280-7 2004 These findings identify a novel p53-independent channel of cross-talk between topoisomerase I inhibitors and Apo2L/TRAIL and suggest that the addition of Apo2L/TRAIL can improve the therapeutic index of irinotecan against both p53-proficient and p53-deficient colorectal cancers, including those that have metastasized to the liver. Irinotecan 203-213 TNF superfamily member 10 Homo sapiens 109-114 15604280-3 2004 Whereas irinotecan-induced up-regulation of the proapoptotic proteins PUMA and Noxa requires p53, we find that irinotecan inhibits Janus kinase 2 (JAK2)-signal transducer and activator of transcription 3 and 5 (STAT3/5) signaling in both p53-proficient and p53-deficient tumor cells. Irinotecan 8-18 tumor protein p53 Homo sapiens 238-241 15604280-7 2004 These findings identify a novel p53-independent channel of cross-talk between topoisomerase I inhibitors and Apo2L/TRAIL and suggest that the addition of Apo2L/TRAIL can improve the therapeutic index of irinotecan against both p53-proficient and p53-deficient colorectal cancers, including those that have metastasized to the liver. Irinotecan 203-213 TNF superfamily member 10 Homo sapiens 115-120 15604280-7 2004 These findings identify a novel p53-independent channel of cross-talk between topoisomerase I inhibitors and Apo2L/TRAIL and suggest that the addition of Apo2L/TRAIL can improve the therapeutic index of irinotecan against both p53-proficient and p53-deficient colorectal cancers, including those that have metastasized to the liver. Irinotecan 203-213 TNF superfamily member 10 Homo sapiens 154-159 15604280-3 2004 Whereas irinotecan-induced up-regulation of the proapoptotic proteins PUMA and Noxa requires p53, we find that irinotecan inhibits Janus kinase 2 (JAK2)-signal transducer and activator of transcription 3 and 5 (STAT3/5) signaling in both p53-proficient and p53-deficient tumor cells. Irinotecan 8-18 tumor protein p53 Homo sapiens 238-241 15604280-7 2004 These findings identify a novel p53-independent channel of cross-talk between topoisomerase I inhibitors and Apo2L/TRAIL and suggest that the addition of Apo2L/TRAIL can improve the therapeutic index of irinotecan against both p53-proficient and p53-deficient colorectal cancers, including those that have metastasized to the liver. Irinotecan 203-213 TNF superfamily member 10 Homo sapiens 160-165 15604280-3 2004 Whereas irinotecan-induced up-regulation of the proapoptotic proteins PUMA and Noxa requires p53, we find that irinotecan inhibits Janus kinase 2 (JAK2)-signal transducer and activator of transcription 3 and 5 (STAT3/5) signaling in both p53-proficient and p53-deficient tumor cells. Irinotecan 111-121 Janus kinase 2 Homo sapiens 147-151 15604280-7 2004 These findings identify a novel p53-independent channel of cross-talk between topoisomerase I inhibitors and Apo2L/TRAIL and suggest that the addition of Apo2L/TRAIL can improve the therapeutic index of irinotecan against both p53-proficient and p53-deficient colorectal cancers, including those that have metastasized to the liver. Irinotecan 203-213 tumor protein p53 Homo sapiens 227-230 15604280-7 2004 These findings identify a novel p53-independent channel of cross-talk between topoisomerase I inhibitors and Apo2L/TRAIL and suggest that the addition of Apo2L/TRAIL can improve the therapeutic index of irinotecan against both p53-proficient and p53-deficient colorectal cancers, including those that have metastasized to the liver. Irinotecan 203-213 tumor protein p53 Homo sapiens 227-230 15604280-3 2004 Whereas irinotecan-induced up-regulation of the proapoptotic proteins PUMA and Noxa requires p53, we find that irinotecan inhibits Janus kinase 2 (JAK2)-signal transducer and activator of transcription 3 and 5 (STAT3/5) signaling in both p53-proficient and p53-deficient tumor cells. Irinotecan 111-121 signal transducer and activator of transcription 3 Homo sapiens 211-218 15604280-3 2004 Whereas irinotecan-induced up-regulation of the proapoptotic proteins PUMA and Noxa requires p53, we find that irinotecan inhibits Janus kinase 2 (JAK2)-signal transducer and activator of transcription 3 and 5 (STAT3/5) signaling in both p53-proficient and p53-deficient tumor cells. Irinotecan 111-121 tumor protein p53 Homo sapiens 238-241 15634630-1 2004 BACKGROUND: To establish the recommended dose (RD) of the thymidylate-synthase inhibitor ZD9331 administered with irinotecan (CPT-11) in patients with pretreated metastatic colorectal cancer, and to assess toxicity profile, pharmacokinetics (PK), and anti-tumor activity in a phase I/II open, multicenter, intrapatient chemotherapy dose escalating trial. Irinotecan 114-124 thymidylate synthetase Homo sapiens 58-78 15634630-1 2004 BACKGROUND: To establish the recommended dose (RD) of the thymidylate-synthase inhibitor ZD9331 administered with irinotecan (CPT-11) in patients with pretreated metastatic colorectal cancer, and to assess toxicity profile, pharmacokinetics (PK), and anti-tumor activity in a phase I/II open, multicenter, intrapatient chemotherapy dose escalating trial. Irinotecan 126-132 thymidylate synthetase Homo sapiens 58-78 15574786-2 2004 Here we show for the first time that SN38, the potent active metabolite of irinotecan, induces c-Jun NH(2)-terminal kinase activation, Fas up-regulation, and caspase 8-mediated apoptosis in multiple myeloma (MM) cells. Irinotecan 75-85 caspase 8 Homo sapiens 158-167 15592324-0 2004 Pharmacogenetics of human carboxylesterase 2, an enzyme involved in the activation of irinotecan into SN-38. Irinotecan 86-96 carboxylesterase 2 Homo sapiens 26-44 20368829-0 2004 Responsiveness of CPT-11 in respect to hMLH1 and hMSH2 protein expression in the primary colorectal cancer. Irinotecan 18-24 mutL homolog 1 Homo sapiens 39-44 20368829-0 2004 Responsiveness of CPT-11 in respect to hMLH1 and hMSH2 protein expression in the primary colorectal cancer. Irinotecan 18-24 mutS homolog 2 Homo sapiens 49-54 20368829-1 2004 PURPOSE: The aim of this study was to evaluate the responsiveness to CPT-11 with respect to hMLH1 and hMSH2 protein expressions in primary colorectal tumors. Irinotecan 69-75 mutL homolog 1 Homo sapiens 92-97 15592324-0 2004 Pharmacogenetics of human carboxylesterase 2, an enzyme involved in the activation of irinotecan into SN-38. Irinotecan 102-107 carboxylesterase 2 Homo sapiens 26-44 20368829-1 2004 PURPOSE: The aim of this study was to evaluate the responsiveness to CPT-11 with respect to hMLH1 and hMSH2 protein expressions in primary colorectal tumors. Irinotecan 69-75 mutS homolog 2 Homo sapiens 102-107 20368829-6 2004 Patients with tumors not expressing hMLH1 showed a significantly better response to CPT-11 (p=0.04). Irinotecan 84-90 mutL homolog 1 Homo sapiens 36-41 15592324-1 2004 PURPOSE: Irinotecan, a drug widely used in the treatment of advanced colorectal cancers, is a prodrug requiring activation to 7-ethyl-10-hydroxycamptothecin (SN-38) by carboxylesterase 2 (hCE2). Irinotecan 9-19 carboxylesterase 2 Homo sapiens 168-186 20368829-10 2004 CONCLUSION: The immunohistochemical determination of loss of hMLH1 and hMSH2 expressions may be used in determining the responsiveness to CPT-11-based chemotherapy. Irinotecan 138-144 mutL homolog 1 Homo sapiens 61-66 20368829-10 2004 CONCLUSION: The immunohistochemical determination of loss of hMLH1 and hMSH2 expressions may be used in determining the responsiveness to CPT-11-based chemotherapy. Irinotecan 138-144 mutS homolog 2 Homo sapiens 71-76 15592324-1 2004 PURPOSE: Irinotecan, a drug widely used in the treatment of advanced colorectal cancers, is a prodrug requiring activation to 7-ethyl-10-hydroxycamptothecin (SN-38) by carboxylesterase 2 (hCE2). Irinotecan 9-19 carboxylesterase 2 Homo sapiens 188-192 15592324-1 2004 PURPOSE: Irinotecan, a drug widely used in the treatment of advanced colorectal cancers, is a prodrug requiring activation to 7-ethyl-10-hydroxycamptothecin (SN-38) by carboxylesterase 2 (hCE2). Irinotecan 126-156 carboxylesterase 2 Homo sapiens 168-186 15592324-1 2004 PURPOSE: Irinotecan, a drug widely used in the treatment of advanced colorectal cancers, is a prodrug requiring activation to 7-ethyl-10-hydroxycamptothecin (SN-38) by carboxylesterase 2 (hCE2). Irinotecan 126-156 carboxylesterase 2 Homo sapiens 188-192 15592324-1 2004 PURPOSE: Irinotecan, a drug widely used in the treatment of advanced colorectal cancers, is a prodrug requiring activation to 7-ethyl-10-hydroxycamptothecin (SN-38) by carboxylesterase 2 (hCE2). Irinotecan 158-163 carboxylesterase 2 Homo sapiens 168-186 15592324-1 2004 PURPOSE: Irinotecan, a drug widely used in the treatment of advanced colorectal cancers, is a prodrug requiring activation to 7-ethyl-10-hydroxycamptothecin (SN-38) by carboxylesterase 2 (hCE2). Irinotecan 158-163 carboxylesterase 2 Homo sapiens 188-192 15592324-10 2004 CONCLUSION: The hCE2 gene presents several polymorphisms, none of which seems to be involved in significant variations in protein activity and, therefore, in irinotecan activation. Irinotecan 158-168 carboxylesterase 2 Homo sapiens 16-20 15536465-0 2004 Inhibition of acetylcholinesterase in patients receiving irinotecan (camptothecin-11). Irinotecan 57-67 acetylcholinesterase (Cartwright blood group) Homo sapiens 14-34 15523087-0 2004 Prediction of irinotecan pharmacokinetics by use of cytochrome P450 3A4 phenotyping probes. Irinotecan 14-24 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 52-71 15523087-2 2004 The response to irinotecan is variable, possibly because of interindividual variation in the expression of the enzymes that metabolize irinotecan, including cytochrome P450 3A4 (CYP3A4) and uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1). Irinotecan 16-26 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 157-176 15523087-2 2004 The response to irinotecan is variable, possibly because of interindividual variation in the expression of the enzymes that metabolize irinotecan, including cytochrome P450 3A4 (CYP3A4) and uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1). Irinotecan 16-26 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 178-184 15523087-2 2004 The response to irinotecan is variable, possibly because of interindividual variation in the expression of the enzymes that metabolize irinotecan, including cytochrome P450 3A4 (CYP3A4) and uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1). Irinotecan 16-26 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 190-237 15523087-2 2004 The response to irinotecan is variable, possibly because of interindividual variation in the expression of the enzymes that metabolize irinotecan, including cytochrome P450 3A4 (CYP3A4) and uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1). Irinotecan 16-26 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 239-245 15523087-2 2004 The response to irinotecan is variable, possibly because of interindividual variation in the expression of the enzymes that metabolize irinotecan, including cytochrome P450 3A4 (CYP3A4) and uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1). Irinotecan 135-145 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 157-176 15523087-2 2004 The response to irinotecan is variable, possibly because of interindividual variation in the expression of the enzymes that metabolize irinotecan, including cytochrome P450 3A4 (CYP3A4) and uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1). Irinotecan 135-145 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 178-184 15523087-2 2004 The response to irinotecan is variable, possibly because of interindividual variation in the expression of the enzymes that metabolize irinotecan, including cytochrome P450 3A4 (CYP3A4) and uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1). Irinotecan 135-145 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 190-237 15523087-2 2004 The response to irinotecan is variable, possibly because of interindividual variation in the expression of the enzymes that metabolize irinotecan, including cytochrome P450 3A4 (CYP3A4) and uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1). Irinotecan 135-145 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 239-245 15523087-12 2004 In addition, the presence of a UGT1A1 variant with a (TA)7 repeat in the promoter (UGT1A1*28) was associated with increased exposure to SN-38 (435 ng x h/mL, 95% confidence interval [CI] = 339 to 531 ng x h/mL in patients who are homozygous for wild-type UGT1A1; 631 ng x h/mL, 95% CI = 499 to 762 ng . Irinotecan 136-141 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 31-37 15523087-12 2004 In addition, the presence of a UGT1A1 variant with a (TA)7 repeat in the promoter (UGT1A1*28) was associated with increased exposure to SN-38 (435 ng x h/mL, 95% confidence interval [CI] = 339 to 531 ng x h/mL in patients who are homozygous for wild-type UGT1A1; 631 ng x h/mL, 95% CI = 499 to 762 ng . Irinotecan 136-141 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 83-89 15523087-12 2004 In addition, the presence of a UGT1A1 variant with a (TA)7 repeat in the promoter (UGT1A1*28) was associated with increased exposure to SN-38 (435 ng x h/mL, 95% confidence interval [CI] = 339 to 531 ng x h/mL in patients who are homozygous for wild-type UGT1A1; 631 ng x h/mL, 95% CI = 499 to 762 ng . Irinotecan 136-141 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 83-89 15523087-14 2004 CONCLUSION: CYP3A4 phenotype, as assessed by midazolam clearance, is statistically significantly associated with irinotecan pharmacokinetics. Irinotecan 113-123 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 12-18 15523087-15 2004 Evaluation of midazolam clearance combined with UGT1A1*28 genotyping may assist with optimization of irinotecan chemotherapy. Irinotecan 101-111 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 48-54 15547701-8 2004 We identified two proto-oncogenes, nuclear receptor of T-cells (NOT) and c-fos, that were up-regulated in doxifluridine- and irinotecan-related regimens but unchanged in the 5-FU-related regimen. Irinotecan 125-135 Fos proto-oncogene, AP-1 transcription factor subunit Homo sapiens 73-78 15547701-10 2004 These results suggest that doxifluridine has a synergistic impact on the therapeutic effect of irinotecan by up-regulating proto-oncogenes such as NOT and c-fos, and thus justify the use of one of the irinotecan and fluoropyrimidine combinations. Irinotecan 95-105 Fos proto-oncogene, AP-1 transcription factor subunit Homo sapiens 155-160 15844664-1 2004 We conducted a phase II study of combination chemotherapy with nedaplatin (NP) with irinotecan (CPT) to determine the effects against unresectable non-small cell lung cancer (NSCLC) and to determine the qualitative and quantitative toxicities of this combination chemotherapy in 70 years or older patients. Irinotecan 84-94 choline phosphotransferase 1 Homo sapiens 96-99 15736428-7 2004 Synergism was observed in ABC-1 and SBC-3 cells when cisplatin was given first, followed by SN-38 (7-ethyl-10-hydroxycamptothecin) and cisplatin. Irinotecan 92-97 ATP binding cassette subfamily A member 1 Homo sapiens 26-31 15736428-7 2004 Synergism was observed in ABC-1 and SBC-3 cells when cisplatin was given first, followed by SN-38 (7-ethyl-10-hydroxycamptothecin) and cisplatin. Irinotecan 99-129 ATP binding cassette subfamily A member 1 Homo sapiens 26-31 15536465-0 2004 Inhibition of acetylcholinesterase in patients receiving irinotecan (camptothecin-11). Irinotecan 69-84 acetylcholinesterase (Cartwright blood group) Homo sapiens 14-34 15492275-3 2004 Breast cancer resistance protein (ABCG2), a transporter that confers resistance to SN-38 (the active metabolite of irinotecan), was readily detected in six of nine xenograft models examined by immunohistochemistry. Irinotecan 83-88 ATP binding cassette subfamily G member 2 (Junior blood group) Mus musculus 34-39 15688606-0 2004 Uridine diphosphoglucuronosyl transferase and methylenetetrahydrofolate reductase polymorphisms as genomic predictors of toxicity and response to irinotecan-, antifolate- and fluoropyrimidine-based chemotherapy. Irinotecan 146-156 methylenetetrahydrofolate reductase Homo sapiens 46-81 15688606-3 2004 In particular, uridine diphosphoglucuronosyl transferase 1A1 (UGT1A1) enzyme is responsible for detoxification of irinotecan active metabolite, SN38. Irinotecan 114-124 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 62-68 15688606-4 2004 A polymorphic structure in the promoter region (UGT1A1*28) may affect irinotecan toxicity and SN38 plasma level. Irinotecan 70-80 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 48-54 15492275-3 2004 Breast cancer resistance protein (ABCG2), a transporter that confers resistance to SN-38 (the active metabolite of irinotecan), was readily detected in six of nine xenograft models examined by immunohistochemistry. Irinotecan 115-125 ATP binding cassette subfamily G member 2 (Junior blood group) Mus musculus 34-39 15492275-4 2004 In vitro gefitinib potently reversed resistance to SN-38 only in a cell line that overexpressed functional ABCG2. Irinotecan 51-56 ATP binding cassette subfamily G member 2 (Junior blood group) Mus musculus 107-112 15492275-9 2004 It is concluded that gefitinib may modulate SN-38 activity at the cellular level to reverse tumor resistance mediated by ABCG2 through inhibiting drug efflux and may be used potentially in humans to modulate the oral bioavailability of a poorly absorbed camptothecin such as irinotecan. Irinotecan 44-49 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 121-126 15354215-0 2004 Thymidylate synthase predictive power is overcome by irinotecan combination therapy with S-1 for gastric cancer. Irinotecan 53-63 thymidylate synthetase Homo sapiens 0-20 15354215-10 2004 TS predictive power was overcome by CPT-11 combination therapy with S-1. Irinotecan 36-42 thymidylate synthetase Homo sapiens 0-2 15374978-8 2004 Conversely, concurrent treatment with SN-38 and UCN-01 resulted in S-phase checkpoint override, an amplified DNA damage response including increased phosphorylation of the DNA double-strand breakage marker H2AX and augmentation of clonogenic inhibition, which was independent of p53. Irinotecan 38-43 H2A.X variant histone Homo sapiens 206-210 15553727-3 2004 Administration of CPT-11 (80 mg/body) by a hepatic arterial infusion inhibited the growth of metastatic liver tumor and decreased serum levels of CEA and CA19-9 for several months without a significant adverse side effect. Irinotecan 18-24 CEA cell adhesion molecule 3 Homo sapiens 146-149 15341677-11 2004 EGFR inhibitors are still experimental in every GI malignancy with the notable exception of cetuximab that is approved for second or third-line therapy of metastatic colorectal cancer, used either alone or in combination with irinotecan (Camptosar, Kalamazoo, Mich). Irinotecan 226-236 epidermal growth factor receptor Homo sapiens 0-4 15469406-3 2004 Although multiple genes may play a role in irinotecan activity, the UDP glycuronosyltransferase 1 family, polypeptide A1 (UGT1A1) enzyme has been strongly associated with toxicity. Irinotecan 43-53 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 122-128 15469406-4 2004 A common dinucleotide repeat polymorphism in the UGT1A1 promoter region (UGT1A1*28) has been correlated with severe toxicity in cancer patients receiving irinotecan-containing therapy. Irinotecan 154-164 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 49-55 15469406-4 2004 A common dinucleotide repeat polymorphism in the UGT1A1 promoter region (UGT1A1*28) has been correlated with severe toxicity in cancer patients receiving irinotecan-containing therapy. Irinotecan 154-164 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 73-79 15292932-1 2004 CPT-11 (irinotecan) is a DNA-topoisomerase I inhibitor with preclinical activity against neuroblastoma (NB) xenografts. Irinotecan 0-6 topoisomerase (DNA) I Mus musculus 25-44 15292932-1 2004 CPT-11 (irinotecan) is a DNA-topoisomerase I inhibitor with preclinical activity against neuroblastoma (NB) xenografts. Irinotecan 8-18 topoisomerase (DNA) I Mus musculus 25-44 15292932-3 2004 IGR-NB8 is an immature NB xenograft with MYCN amplification and 1p deletion, which is sensitive to CPT-11. Irinotecan 99-105 v-myc avian myelocytomatosis viral related oncogene, neuroblastoma derived Mus musculus 41-45 15292935-0 2004 Phase I-II study of irinotecan (CPT-11) plus nedaplatin (254-S) with recombinant human granulocyte colony-stimulating factor support in patients with advanced or recurrent cervical cancer. Irinotecan 20-30 colony stimulating factor 3 Homo sapiens 87-124 15374978-8 2004 Conversely, concurrent treatment with SN-38 and UCN-01 resulted in S-phase checkpoint override, an amplified DNA damage response including increased phosphorylation of the DNA double-strand breakage marker H2AX and augmentation of clonogenic inhibition, which was independent of p53. Irinotecan 38-43 tumor protein p53 Homo sapiens 279-282 15367706-9 2004 Furthermore, P388/BCRP-transplanted mice treated with combination of irinotecan and gefitinib survived significantly longer than those treated with irinotecan alone or gefitinib alone. Irinotecan 69-79 ATP binding cassette subfamily G member 2 (Junior blood group) Mus musculus 18-22 15517893-0 2004 UGT1A10 is responsible for SN-38 glucuronidation and its expression in human lung cancers. Irinotecan 27-32 UDP glucuronosyltransferase family 1 member A10 Homo sapiens 0-7 15517893-2 2004 In order to extend this result to the clinical setting, it is important to elucidate the role of SN-38 glucuronidation by UGT1A isoforms in CPT-11/SN-38 resistance in vivo. Irinotecan 97-102 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 122-127 15517893-2 2004 In order to extend this result to the clinical setting, it is important to elucidate the role of SN-38 glucuronidation by UGT1A isoforms in CPT-11/SN-38 resistance in vivo. Irinotecan 140-146 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 122-127 15517893-2 2004 In order to extend this result to the clinical setting, it is important to elucidate the role of SN-38 glucuronidation by UGT1A isoforms in CPT-11/SN-38 resistance in vivo. Irinotecan 147-152 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 122-127 15517893-3 2004 MATERIALS AND METHODS: We examined SN-38 glucuronidation activity in COS-7 cells transfected with full-length cDNAs for human UGT isoforms (UGT1A1, UGT1A3, UGT1A6 and UGT1A10). Irinotecan 35-40 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 126-129 15517893-5 2004 RESULTS: Our HPLC assay results showed that both UGT1A1 and UGT1A10 are responsible for SN-38 glucuronidation. Irinotecan 88-93 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 49-55 15517893-5 2004 RESULTS: Our HPLC assay results showed that both UGT1A1 and UGT1A10 are responsible for SN-38 glucuronidation. Irinotecan 88-93 UDP glucuronosyltransferase family 1 member A10 Homo sapiens 60-67 15517893-6 2004 The levels of UGT1A1 and UGT1A10 expression in a CPT-11/SN-38-resistant cell line were increased compared to levels in the parent cell line. Irinotecan 49-55 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 14-20 15517893-6 2004 The levels of UGT1A1 and UGT1A10 expression in a CPT-11/SN-38-resistant cell line were increased compared to levels in the parent cell line. Irinotecan 49-55 UDP glucuronosyltransferase family 1 member A10 Homo sapiens 25-32 15517893-6 2004 The levels of UGT1A1 and UGT1A10 expression in a CPT-11/SN-38-resistant cell line were increased compared to levels in the parent cell line. Irinotecan 56-61 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 14-20 15517893-6 2004 The levels of UGT1A1 and UGT1A10 expression in a CPT-11/SN-38-resistant cell line were increased compared to levels in the parent cell line. Irinotecan 56-61 UDP glucuronosyltransferase family 1 member A10 Homo sapiens 25-32 15517893-9 2004 Our results suggest that not only UGT 1A1, but also UGT 1A10, plays an important role in detoxifying CPT-11/SN-38, leading to resistance to CPT-11/SN-38 in lung cancer. Irinotecan 101-107 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 34-41 15517893-9 2004 Our results suggest that not only UGT 1A1, but also UGT 1A10, plays an important role in detoxifying CPT-11/SN-38, leading to resistance to CPT-11/SN-38 in lung cancer. Irinotecan 101-107 UDP glucuronosyltransferase family 1 member A10 Homo sapiens 52-60 15517893-9 2004 Our results suggest that not only UGT 1A1, but also UGT 1A10, plays an important role in detoxifying CPT-11/SN-38, leading to resistance to CPT-11/SN-38 in lung cancer. Irinotecan 108-113 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 34-41 15517893-9 2004 Our results suggest that not only UGT 1A1, but also UGT 1A10, plays an important role in detoxifying CPT-11/SN-38, leading to resistance to CPT-11/SN-38 in lung cancer. Irinotecan 108-113 UDP glucuronosyltransferase family 1 member A10 Homo sapiens 52-60 15517893-9 2004 Our results suggest that not only UGT 1A1, but also UGT 1A10, plays an important role in detoxifying CPT-11/SN-38, leading to resistance to CPT-11/SN-38 in lung cancer. Irinotecan 140-146 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 34-41 15517893-9 2004 Our results suggest that not only UGT 1A1, but also UGT 1A10, plays an important role in detoxifying CPT-11/SN-38, leading to resistance to CPT-11/SN-38 in lung cancer. Irinotecan 140-146 UDP glucuronosyltransferase family 1 member A10 Homo sapiens 52-60 15517893-9 2004 Our results suggest that not only UGT 1A1, but also UGT 1A10, plays an important role in detoxifying CPT-11/SN-38, leading to resistance to CPT-11/SN-38 in lung cancer. Irinotecan 147-152 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 34-41 15517893-9 2004 Our results suggest that not only UGT 1A1, but also UGT 1A10, plays an important role in detoxifying CPT-11/SN-38, leading to resistance to CPT-11/SN-38 in lung cancer. Irinotecan 147-152 UDP glucuronosyltransferase family 1 member A10 Homo sapiens 52-60 15355921-0 2004 ABCG2 pharmacogenetics: ethnic differences in allele frequency and assessment of influence on irinotecan disposition. Irinotecan 94-104 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 0-5 15355921-1 2004 PURPOSE: The ATP-binding cassette transporter ABCG2 (breast cancer resistance protein) is an efflux protein that plays a role in host detoxification of various xenobiotic substrates, including the irinotecan metabolite 7- ethyl-10-hydroxycamptothecin (SN-38). Irinotecan 197-207 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 46-51 15355921-1 2004 PURPOSE: The ATP-binding cassette transporter ABCG2 (breast cancer resistance protein) is an efflux protein that plays a role in host detoxification of various xenobiotic substrates, including the irinotecan metabolite 7- ethyl-10-hydroxycamptothecin (SN-38). Irinotecan 197-207 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 53-85 15355921-1 2004 PURPOSE: The ATP-binding cassette transporter ABCG2 (breast cancer resistance protein) is an efflux protein that plays a role in host detoxification of various xenobiotic substrates, including the irinotecan metabolite 7- ethyl-10-hydroxycamptothecin (SN-38). Irinotecan 219-250 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 46-51 15355921-1 2004 PURPOSE: The ATP-binding cassette transporter ABCG2 (breast cancer resistance protein) is an efflux protein that plays a role in host detoxification of various xenobiotic substrates, including the irinotecan metabolite 7- ethyl-10-hydroxycamptothecin (SN-38). Irinotecan 219-250 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 53-85 15355921-1 2004 PURPOSE: The ATP-binding cassette transporter ABCG2 (breast cancer resistance protein) is an efflux protein that plays a role in host detoxification of various xenobiotic substrates, including the irinotecan metabolite 7- ethyl-10-hydroxycamptothecin (SN-38). Irinotecan 252-257 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 46-51 15355921-1 2004 PURPOSE: The ATP-binding cassette transporter ABCG2 (breast cancer resistance protein) is an efflux protein that plays a role in host detoxification of various xenobiotic substrates, including the irinotecan metabolite 7- ethyl-10-hydroxycamptothecin (SN-38). Irinotecan 252-257 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 53-85 15355921-3 2004 The aim of this study was to evaluate the ethnic distribution and potential functional consequence of the ABCG2 421C>A genotype in cancer patients treated with irinotecan. Irinotecan 163-173 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 106-111 15355921-4 2004 EXPERIMENTAL DESIGN: ABCG2 genotyping was performed using Pyrosequencing on DNA from 88 American Caucasians, 94 African Americans, 938 Africans, and 95 Han Chinese, as well as in 84 European Caucasian patients treated with irinotecan undergoing additional blood sampling for pharmacokinetic studies. Irinotecan 223-233 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 21-26 15355921-9 2004 CONCLUSIONS: The ABCG2 421C>A polymorphism appears to play a limited role in the disposition of irinotecan in European Caucasians. Irinotecan 99-109 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 17-22 19780248-7 2004 Cetuximab (an epidermal growth factor receptor antibody) plus irinotecan yields an increased overall response in patients with irinotecan-refractory colorectal disease. Irinotecan 127-137 epidermal growth factor receptor Homo sapiens 14-46 15367706-1 2004 Breast cancer resistance protein (BCRP) is an ATP binding cassette transporter that confers resistance to a series of anticancer agents such as 7-ethyl-10-hydroxycamptothecin (SN-38), topotecan, and mitoxantrone. Irinotecan 144-174 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 0-32 15367706-1 2004 Breast cancer resistance protein (BCRP) is an ATP binding cassette transporter that confers resistance to a series of anticancer agents such as 7-ethyl-10-hydroxycamptothecin (SN-38), topotecan, and mitoxantrone. Irinotecan 144-174 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 34-38 15367706-1 2004 Breast cancer resistance protein (BCRP) is an ATP binding cassette transporter that confers resistance to a series of anticancer agents such as 7-ethyl-10-hydroxycamptothecin (SN-38), topotecan, and mitoxantrone. Irinotecan 176-181 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 0-32 15367706-1 2004 Breast cancer resistance protein (BCRP) is an ATP binding cassette transporter that confers resistance to a series of anticancer agents such as 7-ethyl-10-hydroxycamptothecin (SN-38), topotecan, and mitoxantrone. Irinotecan 176-181 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 34-38 15367706-5 2004 Gefitinib reversed SN-38 resistance in BCRP-transduced human myelogenous leukemia K562 (K562/BCRP) or BCRP-transduced murine lymphocytic leukemia P388 (P388/BCRP) cells but not in these parental cells. Irinotecan 19-24 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 39-43 15367706-6 2004 In addition, gefitinib sensitized human colon cancer HT-29 cells, which endogenously express BCRP, to SN-38. Irinotecan 102-107 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 93-97 15280927-1 2004 SN-38 is the active metabolite of irinotecan and it is metabolised through conjugation by uridine diphosphate glucuronosyl transferase (UGT1A1). Irinotecan 0-5 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 136-142 15280927-1 2004 SN-38 is the active metabolite of irinotecan and it is metabolised through conjugation by uridine diphosphate glucuronosyl transferase (UGT1A1). Irinotecan 34-44 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 136-142 15280927-2 2004 The major toxicity of irinotecan therapy is diarrhoea, which has been related to the enzymatic activity of UGT1A1. Irinotecan 22-32 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 107-113 15280927-13 2004 The role of the UGT1A1 genotype as a predictor of toxicity in cancer patients receiving irinotecan demands the performance of a randomized trial to ascertain whether genotype-adjusted dosages of the drug can help to establish safe and effective doses not only for patients with the UGT1A1(*)28 homozygous genotype but also for those with the most common UGT1A1 6/6 or 6/7 genotype. Irinotecan 88-98 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 16-22 15286088-0 2004 Influence of genetic variants in UGT1A1 and UGT1A9 on the in vivo glucuronidation of SN-38. Irinotecan 85-90 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-39 15297419-0 2004 Relevance of different UGT1A1 polymorphisms in irinotecan-induced toxicity: a molecular and clinical study of 75 patients. Irinotecan 47-57 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 23-29 15297419-1 2004 PURPOSE: We wanted to assess polymorphisms in the uridine diphosphoglucuronosyl transferase 1A1 (UGT 1A1) gene: the TATA box polymorphism and UGT 1A1 G71R and Y486D mutations in the coding sequence, the main mutations characterizing Gilbert"s syndrome, as predictors of severe toxic event occurrence after irinotecan (CPT-11) administration. Irinotecan 306-316 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 50-95 15297419-1 2004 PURPOSE: We wanted to assess polymorphisms in the uridine diphosphoglucuronosyl transferase 1A1 (UGT 1A1) gene: the TATA box polymorphism and UGT 1A1 G71R and Y486D mutations in the coding sequence, the main mutations characterizing Gilbert"s syndrome, as predictors of severe toxic event occurrence after irinotecan (CPT-11) administration. Irinotecan 306-316 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 97-104 15297419-1 2004 PURPOSE: We wanted to assess polymorphisms in the uridine diphosphoglucuronosyl transferase 1A1 (UGT 1A1) gene: the TATA box polymorphism and UGT 1A1 G71R and Y486D mutations in the coding sequence, the main mutations characterizing Gilbert"s syndrome, as predictors of severe toxic event occurrence after irinotecan (CPT-11) administration. Irinotecan 318-324 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 50-95 15297419-1 2004 PURPOSE: We wanted to assess polymorphisms in the uridine diphosphoglucuronosyl transferase 1A1 (UGT 1A1) gene: the TATA box polymorphism and UGT 1A1 G71R and Y486D mutations in the coding sequence, the main mutations characterizing Gilbert"s syndrome, as predictors of severe toxic event occurrence after irinotecan (CPT-11) administration. Irinotecan 318-324 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 97-104 15275881-2 2004 Inhibition of NF-kappaB by adenoviral delivery of an IkappaBalpha superrepressor (Ad.IkappaBalpha-SR) should potentiate 5-fluorouracil (5-FU) and irinotecan chemotherapy in gastric cancer cells. Irinotecan 146-156 nuclear factor kappa B subunit 1 Homo sapiens 14-23 15275881-2 2004 Inhibition of NF-kappaB by adenoviral delivery of an IkappaBalpha superrepressor (Ad.IkappaBalpha-SR) should potentiate 5-fluorouracil (5-FU) and irinotecan chemotherapy in gastric cancer cells. Irinotecan 146-156 NFKB inhibitor alpha Homo sapiens 53-65 15275881-2 2004 Inhibition of NF-kappaB by adenoviral delivery of an IkappaBalpha superrepressor (Ad.IkappaBalpha-SR) should potentiate 5-fluorouracil (5-FU) and irinotecan chemotherapy in gastric cancer cells. Irinotecan 146-156 NFKB inhibitor alpha Homo sapiens 85-97 15275881-8 2004 RESULTS: 5-FU and SN-38 significantly induced NF-kappaB activation as measured by luciferase reporter assay (p < 0.001). Irinotecan 18-23 nuclear factor kappa B subunit 1 Homo sapiens 46-55 15275881-10 2004 In AGS cells, pretreatment with Ad.IkappaBalpha-SR followed by 5-FU (0.005 mmol/L) or SN-38 (10 ng/mL) led to increased growth inhibition of 13% and 59%, respectively (p < 0.001). Irinotecan 86-91 NFKB inhibitor alpha Homo sapiens 35-47 15286088-8 2004 The median AUC ratio of SN-38G to SN-38 was significantly reduced in carriers of the variant UGT1A1(*)28 allele (7.00 [Wt] vs. 6.26 [Het] vs. 2.51 [Var]; p =.022). Irinotecan 24-29 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-99 15286088-8 2004 The median AUC ratio of SN-38G to SN-38 was significantly reduced in carriers of the variant UGT1A1(*)28 allele (7.00 [Wt] vs. 6.26 [Het] vs. 2.51 [Var]; p =.022). Irinotecan 34-39 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-99 15286088-9 2004 It is concluded that UGT1A9 functional variants are rare in Caucasians and likely to be clinically insignificant in irinotecan regimens. Irinotecan 116-126 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 21-27 15286088-10 2004 Screening for the UGT1A1(*)28 polymorphism may identify patients with altered SN-38 pharmacokinetics. Irinotecan 78-83 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 18-24 15286088-0 2004 Influence of genetic variants in UGT1A1 and UGT1A9 on the in vivo glucuronidation of SN-38. Irinotecan 85-90 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 44-50 15286088-1 2004 The uridine diphosphate glucuronosyltransferase (UGT) 1A1 and 1A9 isoforms are involved in the phase II biotransformation of the irinotecan metabolite, SN-38. Irinotecan 129-139 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-65 15286088-1 2004 The uridine diphosphate glucuronosyltransferase (UGT) 1A1 and 1A9 isoforms are involved in the phase II biotransformation of the irinotecan metabolite, SN-38. Irinotecan 152-157 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-65 15286088-3 2004 The aim of this study was to evaluate the functional consequence of the UGT1A1(TA)(7)TAA (UGT1A1(*)28), UGT1A9 766G>A (D256N; UGT1A9(*)5), and UGT1A9 98T>C (M33T; UGT1A9(*)3) variants in Caucasian patients treated with irinotecan. Irinotecan 225-235 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 72-78 15286088-3 2004 The aim of this study was to evaluate the functional consequence of the UGT1A1(TA)(7)TAA (UGT1A1(*)28), UGT1A9 766G>A (D256N; UGT1A9(*)5), and UGT1A9 98T>C (M33T; UGT1A9(*)3) variants in Caucasian patients treated with irinotecan. Irinotecan 225-235 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 104-110 15286088-3 2004 The aim of this study was to evaluate the functional consequence of the UGT1A1(TA)(7)TAA (UGT1A1(*)28), UGT1A9 766G>A (D256N; UGT1A9(*)5), and UGT1A9 98T>C (M33T; UGT1A9(*)3) variants in Caucasian patients treated with irinotecan. Irinotecan 225-235 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 129-135 15286088-3 2004 The aim of this study was to evaluate the functional consequence of the UGT1A1(TA)(7)TAA (UGT1A1(*)28), UGT1A9 766G>A (D256N; UGT1A9(*)5), and UGT1A9 98T>C (M33T; UGT1A9(*)3) variants in Caucasian patients treated with irinotecan. Irinotecan 225-235 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 129-135 15286088-3 2004 The aim of this study was to evaluate the functional consequence of the UGT1A1(TA)(7)TAA (UGT1A1(*)28), UGT1A9 766G>A (D256N; UGT1A9(*)5), and UGT1A9 98T>C (M33T; UGT1A9(*)3) variants in Caucasian patients treated with irinotecan. Irinotecan 225-235 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 129-135 15075385-0 2004 ABCG2 mediates differential resistance to SN-38 (7-ethyl-10-hydroxycamptothecin) and homocamptothecins. Irinotecan 42-47 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 0-5 15075385-0 2004 ABCG2 mediates differential resistance to SN-38 (7-ethyl-10-hydroxycamptothecin) and homocamptothecins. Irinotecan 49-79 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 0-5 15075385-5 2004 However, the resistance of three selected cell lines overexpressing wild-type or mutant ABCG2 to homocamptothecin or BN80915 was less than resistance to SN-38 (7-ethyl-10-hydroxycamptothecin), indicating that both the seven-membered E-ring present in homocamptothecin and the A- and B-ring modifications present in SN-38 are involved in substrate recognition by ABCG2. Irinotecan 315-320 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 88-93 15075385-6 2004 HEK-293 cells transfected with vectors encoding wild-type or mutant ABCG2 were found to be less resistant to both homocamptothecins than to SN-38. Irinotecan 140-145 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 68-73 15075385-7 2004 However, transfectants overexpressing mutant ABCG2 had relative resistance values for homocamptothecin and BN80915 4- to 14-fold higher than cells expressing wild-type ABCG2, suggesting that the gain of function resulting from mutation at amino acid 482, although not affecting SN-38, extends to the homocamptothecins. Irinotecan 278-283 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 45-50 15075385-8 2004 Resistance was reversed by the ABCG2 inhibitor fumitremorgin C. BN80915 was 17-fold more potent than SN-38 in wild-type ABCG2-transfected cells, suggesting that BN80915 has the potential to overcome ABCG2-related resistance to SN-38, the active metabolite of CPT-11 (irinotecan). Irinotecan 101-106 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 31-36 15075385-8 2004 Resistance was reversed by the ABCG2 inhibitor fumitremorgin C. BN80915 was 17-fold more potent than SN-38 in wild-type ABCG2-transfected cells, suggesting that BN80915 has the potential to overcome ABCG2-related resistance to SN-38, the active metabolite of CPT-11 (irinotecan). Irinotecan 227-232 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 31-36 15075385-8 2004 Resistance was reversed by the ABCG2 inhibitor fumitremorgin C. BN80915 was 17-fold more potent than SN-38 in wild-type ABCG2-transfected cells, suggesting that BN80915 has the potential to overcome ABCG2-related resistance to SN-38, the active metabolite of CPT-11 (irinotecan). Irinotecan 259-265 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 31-36 15075385-8 2004 Resistance was reversed by the ABCG2 inhibitor fumitremorgin C. BN80915 was 17-fold more potent than SN-38 in wild-type ABCG2-transfected cells, suggesting that BN80915 has the potential to overcome ABCG2-related resistance to SN-38, the active metabolite of CPT-11 (irinotecan). Irinotecan 267-277 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 31-36 15256434-4 2004 Compared with control cells, metallothionein 1X (MT1X)-transfected AGS cells showed a 1.4-fold higher irinotecan IC(50) by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and tended to form more colonies. Irinotecan 102-112 metallothionein 1X Homo sapiens 29-47 15254716-1 2004 (Uridino-diphosphate)glucuronosyl-transferase enzyme 1A1 isoform (UGT1A1) is involved in glucuronidation of antineoplastic drugs such as SN38, the active metabolite of irinotecan, as well as estrogens and their metabolites. Irinotecan 168-178 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 66-72 15170677-1 2004 Irinotecan (7-ethyl-10-[4-(1-piperidino)-1-piperidino]-carbonyloxycamptothecin; CPT-11) is a widely used potent antitumor drug that inhibits mammalian DNA topoisomerase I (Topo I); however, overexpression of ABCG2 (BCRP/MXR/ABCP) can confer cancer cell resistance to SN-38, the active form of CPT-11. Irinotecan 0-10 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 208-213 15170677-1 2004 Irinotecan (7-ethyl-10-[4-(1-piperidino)-1-piperidino]-carbonyloxycamptothecin; CPT-11) is a widely used potent antitumor drug that inhibits mammalian DNA topoisomerase I (Topo I); however, overexpression of ABCG2 (BCRP/MXR/ABCP) can confer cancer cell resistance to SN-38, the active form of CPT-11. Irinotecan 0-10 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 215-219 15170677-1 2004 Irinotecan (7-ethyl-10-[4-(1-piperidino)-1-piperidino]-carbonyloxycamptothecin; CPT-11) is a widely used potent antitumor drug that inhibits mammalian DNA topoisomerase I (Topo I); however, overexpression of ABCG2 (BCRP/MXR/ABCP) can confer cancer cell resistance to SN-38, the active form of CPT-11. Irinotecan 0-10 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 220-223 15170677-1 2004 Irinotecan (7-ethyl-10-[4-(1-piperidino)-1-piperidino]-carbonyloxycamptothecin; CPT-11) is a widely used potent antitumor drug that inhibits mammalian DNA topoisomerase I (Topo I); however, overexpression of ABCG2 (BCRP/MXR/ABCP) can confer cancer cell resistance to SN-38, the active form of CPT-11. Irinotecan 0-10 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 224-228 15170677-1 2004 Irinotecan (7-ethyl-10-[4-(1-piperidino)-1-piperidino]-carbonyloxycamptothecin; CPT-11) is a widely used potent antitumor drug that inhibits mammalian DNA topoisomerase I (Topo I); however, overexpression of ABCG2 (BCRP/MXR/ABCP) can confer cancer cell resistance to SN-38, the active form of CPT-11. Irinotecan 80-86 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 208-213 15170677-1 2004 Irinotecan (7-ethyl-10-[4-(1-piperidino)-1-piperidino]-carbonyloxycamptothecin; CPT-11) is a widely used potent antitumor drug that inhibits mammalian DNA topoisomerase I (Topo I); however, overexpression of ABCG2 (BCRP/MXR/ABCP) can confer cancer cell resistance to SN-38, the active form of CPT-11. Irinotecan 80-86 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 215-219 15170677-1 2004 Irinotecan (7-ethyl-10-[4-(1-piperidino)-1-piperidino]-carbonyloxycamptothecin; CPT-11) is a widely used potent antitumor drug that inhibits mammalian DNA topoisomerase I (Topo I); however, overexpression of ABCG2 (BCRP/MXR/ABCP) can confer cancer cell resistance to SN-38, the active form of CPT-11. Irinotecan 80-86 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 220-223 15170677-1 2004 Irinotecan (7-ethyl-10-[4-(1-piperidino)-1-piperidino]-carbonyloxycamptothecin; CPT-11) is a widely used potent antitumor drug that inhibits mammalian DNA topoisomerase I (Topo I); however, overexpression of ABCG2 (BCRP/MXR/ABCP) can confer cancer cell resistance to SN-38, the active form of CPT-11. Irinotecan 80-86 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 224-228 15170677-2 2004 We have recently demonstrated that plasma membrane vesicles prepared from ABCG2-overexpressing PC-6/SN2-5H cells transported SN-38 and its glucuronide conjugate in an ATP-dependent manner (Nakatomi et al., Biochem Biophys Res Commun 2001;288:827-32). Irinotecan 125-130 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 74-79 15170677-2 2004 We have recently demonstrated that plasma membrane vesicles prepared from ABCG2-overexpressing PC-6/SN2-5H cells transported SN-38 and its glucuronide conjugate in an ATP-dependent manner (Nakatomi et al., Biochem Biophys Res Commun 2001;288:827-32). Irinotecan 125-130 proprotein convertase subtilisin/kexin type 5 Homo sapiens 95-99 15170677-5 2004 Their antitumor activities in the SN-38-resistant PC-6/SN2-5H2 cell line greatly varied, however, being correlated with intracellular accumulation levels. Irinotecan 34-39 proprotein convertase subtilisin/kexin type 5 Homo sapiens 50-54 15299073-5 2004 A secondary goal was to develop molecules that specifically inhibit activation of CPT-11 by a rabbit liver carboxylesterase (rCE). Irinotecan 82-88 liver carboxylesterase 1 Oryctolagus cuniculus 101-123 15254685-0 2004 Cisplatin enhances the p53-independent apoptosis induced by a topoisomerase I inhibitor (CPT-11) in the lens epithelial tumors in transgenic mice. Irinotecan 89-95 transformation related protein 53, pseudogene Mus musculus 23-26 15254685-6 2004 In addition, it was found that Cisplatin augmented CPT-11-induced p53-independent apoptosis in p53-deficient alphaT3 mice. Irinotecan 51-57 transformation related protein 53, pseudogene Mus musculus 66-69 15254685-6 2004 In addition, it was found that Cisplatin augmented CPT-11-induced p53-independent apoptosis in p53-deficient alphaT3 mice. Irinotecan 51-57 transformation related protein 53, pseudogene Mus musculus 95-98 15256434-4 2004 Compared with control cells, metallothionein 1X (MT1X)-transfected AGS cells showed a 1.4-fold higher irinotecan IC(50) by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and tended to form more colonies. Irinotecan 102-112 metallothionein 1X Homo sapiens 49-53 15205350-4 2004 In this study, we show that phytoestrogens/flavonoids, such as genistein, naringenin, acacetin, and kaempferol, potentiated the cytotoxicity of SN-38 and mitoxantrone in BCRP-transduced K562 (K562/BCRP) cells. Irinotecan 144-149 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 170-174 15160289-1 2004 PURPOSE: To evaluate the ability of D-saccharic acid 1.4-lactone (SAL), a beta-glucuronidase inhibitor, to prevent irinotecan hydrochloride (CPT-11) from inducing mucosal damage as a cause of diarrhea in rats. Irinotecan 141-147 glucuronidase, beta Rattus norvegicus 74-92 15160289-11 2004 CONCLUSIONS: The beta-glucuronidase inhibitor SAL is able to significantly reduce CPT-11-induced mucosal damage in the small intestine of rats. Irinotecan 82-88 glucuronidase, beta Rattus norvegicus 17-35 15213307-3 2004 Activated MSC increases chk2 phosphorylation at threonine-68 induced by SN-38, with no significant effect on chk1 phosphorylation. Irinotecan 72-77 checkpoint kinase 2 Homo sapiens 24-28 15213307-10 2004 The observed down-regulation of DNA replication proteins cdc6, MCM2, and cdc25A after exposure to SN-38 with MSC further indicates a relationship between drug response and DNA damage. Irinotecan 98-103 cell division cycle 6 Homo sapiens 57-61 15213307-10 2004 The observed down-regulation of DNA replication proteins cdc6, MCM2, and cdc25A after exposure to SN-38 with MSC further indicates a relationship between drug response and DNA damage. Irinotecan 98-103 minichromosome maintenance complex component 2 Homo sapiens 63-67 15213307-10 2004 The observed down-regulation of DNA replication proteins cdc6, MCM2, and cdc25A after exposure to SN-38 with MSC further indicates a relationship between drug response and DNA damage. Irinotecan 98-103 cell division cycle 25A Homo sapiens 73-79 15213307-11 2004 Exposure to SN-38 with MSC resulted in a significant increase of poly(ADP-ribose) polymerasecleavage and caspase 3 activation. Irinotecan 12-17 caspase 3 Homo sapiens 105-114 15183125-9 2004 Interestingly, SN-38 was able to activate EGFR and its signal transduction pathway. Irinotecan 15-20 epidermal growth factor receptor Homo sapiens 42-46 15213307-0 2004 Enhanced 7-ethyl-10-hydroxycamptothecin (SN-38) lethality by methylselenocysteine is associated with Chk2 phosphorylation at threonine-68 and down-regulation of Cdc6 expression. Irinotecan 9-39 checkpoint kinase 2 Homo sapiens 101-105 15213307-0 2004 Enhanced 7-ethyl-10-hydroxycamptothecin (SN-38) lethality by methylselenocysteine is associated with Chk2 phosphorylation at threonine-68 and down-regulation of Cdc6 expression. Irinotecan 9-39 cell division cycle 6 Homo sapiens 161-165 15213307-0 2004 Enhanced 7-ethyl-10-hydroxycamptothecin (SN-38) lethality by methylselenocysteine is associated with Chk2 phosphorylation at threonine-68 and down-regulation of Cdc6 expression. Irinotecan 41-46 checkpoint kinase 2 Homo sapiens 101-105 15213307-0 2004 Enhanced 7-ethyl-10-hydroxycamptothecin (SN-38) lethality by methylselenocysteine is associated with Chk2 phosphorylation at threonine-68 and down-regulation of Cdc6 expression. Irinotecan 41-46 cell division cycle 6 Homo sapiens 161-165 15213307-2 2004 We found that methioninase-activated MSC potentiates 7-ethyl-10-hydroxycamptothecin (SN-38)-induced cell lethality in vitro in the p53-defective human head and neck carcinoma A253 cells. Irinotecan 53-83 tumor protein p53 Homo sapiens 131-134 15213307-2 2004 We found that methioninase-activated MSC potentiates 7-ethyl-10-hydroxycamptothecin (SN-38)-induced cell lethality in vitro in the p53-defective human head and neck carcinoma A253 cells. Irinotecan 85-90 tumor protein p53 Homo sapiens 131-134 15205350-1 2004 Breast cancer resistance protein (BCRP), also called ABCG2, confers resistance to anticancer agents such as 7-ethyl-10-hydroxycamptothecin (SN-38), mitoxantrone, and topotecan. Irinotecan 108-138 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 0-32 15205350-1 2004 Breast cancer resistance protein (BCRP), also called ABCG2, confers resistance to anticancer agents such as 7-ethyl-10-hydroxycamptothecin (SN-38), mitoxantrone, and topotecan. Irinotecan 108-138 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 34-38 15205350-1 2004 Breast cancer resistance protein (BCRP), also called ABCG2, confers resistance to anticancer agents such as 7-ethyl-10-hydroxycamptothecin (SN-38), mitoxantrone, and topotecan. Irinotecan 108-138 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 53-58 15205350-1 2004 Breast cancer resistance protein (BCRP), also called ABCG2, confers resistance to anticancer agents such as 7-ethyl-10-hydroxycamptothecin (SN-38), mitoxantrone, and topotecan. Irinotecan 140-145 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 0-32 15205350-1 2004 Breast cancer resistance protein (BCRP), also called ABCG2, confers resistance to anticancer agents such as 7-ethyl-10-hydroxycamptothecin (SN-38), mitoxantrone, and topotecan. Irinotecan 140-145 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 34-38 15205350-1 2004 Breast cancer resistance protein (BCRP), also called ABCG2, confers resistance to anticancer agents such as 7-ethyl-10-hydroxycamptothecin (SN-38), mitoxantrone, and topotecan. Irinotecan 140-145 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 53-58 15205350-4 2004 In this study, we show that phytoestrogens/flavonoids, such as genistein, naringenin, acacetin, and kaempferol, potentiated the cytotoxicity of SN-38 and mitoxantrone in BCRP-transduced K562 (K562/BCRP) cells. Irinotecan 144-149 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 197-201 15132777-0 2004 The expressions of p21 and pRB may be good indicators for the sensitivity of esophageal squamous cell cancers to CPT-11: Cell proliferation activity correlates with the effect of CPT-11. Irinotecan 113-119 H3 histone pseudogene 16 Homo sapiens 19-22 15179405-0 2004 UGT1A1 haplotypes associated with reduced glucuronidation and increased serum bilirubin in irinotecan-administered Japanese patients with cancer. Irinotecan 91-101 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 15027118-10 2004 Furthermore, we show, for the first time in clinical samples, that the ABCG2 mRNA content in hepatic metastases is higher after an irinotecan-based chemotherapy than in irinotecan-naive metastases. Irinotecan 131-141 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 71-76 15027118-10 2004 Furthermore, we show, for the first time in clinical samples, that the ABCG2 mRNA content in hepatic metastases is higher after an irinotecan-based chemotherapy than in irinotecan-naive metastases. Irinotecan 169-179 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 71-76 15027118-11 2004 In conclusion, this study supports the potential involvement of ABCG2 in the development of irinotecan resistance in vivo. Irinotecan 92-102 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 64-69 14760509-2 2004 Given the high frequency of K-Ras mutations in malignancies commonly treated with irinotecan, the broad preclinical antiproliferative activity of R115777 and its largely non-overlapping toxicity profile with irinotecan, this phase I study of the combination of R115777 and irinotecan in patients with advanced cancer was undertaken. Irinotecan 82-92 KRAS proto-oncogene, GTPase Homo sapiens 28-33 15161687-0 2004 Enhanced chemosensitivity to irinotecan by RNA interference-mediated down-regulation of the nuclear factor-kappaB p65 subunit. Irinotecan 29-39 RELA proto-oncogene, NF-kB subunit Homo sapiens 114-117 15161687-2 2004 This is based on the fact that a variety of chemotherapy agents such as CPT-11 activate NF-kappaB to result in the expression of genes such as c-IAP1 and c-IAP2 that might be responsible for the inhibition of chemotherapy-induced apoptosis. Irinotecan 72-78 baculoviral IAP repeat containing 2 Homo sapiens 143-149 15161687-2 2004 This is based on the fact that a variety of chemotherapy agents such as CPT-11 activate NF-kappaB to result in the expression of genes such as c-IAP1 and c-IAP2 that might be responsible for the inhibition of chemotherapy-induced apoptosis. Irinotecan 72-78 baculoviral IAP repeat containing 3 Homo sapiens 154-160 15161687-4 2004 Reduction of endogenous p65 by siRNA treatment significantly impaired CPT-11-mediated NF-kappaB activation, enhanced apoptosis, and reduced colony formation in soft agar. Irinotecan 70-76 RELA proto-oncogene, NF-kB subunit Homo sapiens 24-27 15027118-0 2004 ABCG2 overexpression in colon cancer cells resistant to SN38 and in irinotecan-treated metastases. Irinotecan 68-78 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 0-5 15027118-3 2004 The aim of the present study was to determine the involvement of ABCG2 in resistance to SN38 (the active metabolite of irinotecan) in colorectal cancer. Irinotecan 119-129 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 65-70 15132777-0 2004 The expressions of p21 and pRB may be good indicators for the sensitivity of esophageal squamous cell cancers to CPT-11: Cell proliferation activity correlates with the effect of CPT-11. Irinotecan 113-119 RB transcriptional corepressor 1 Homo sapiens 27-30 15132777-0 2004 The expressions of p21 and pRB may be good indicators for the sensitivity of esophageal squamous cell cancers to CPT-11: Cell proliferation activity correlates with the effect of CPT-11. Irinotecan 179-185 H3 histone pseudogene 16 Homo sapiens 19-22 15132777-4 2004 Among these proteins, the expression levels of p21 and pRB showed significant differences that were associated with the IC50 values for CPT-11 (P = 0.0339 and P = 0.0109, respectively). Irinotecan 136-142 H3 histone pseudogene 16 Homo sapiens 47-50 15132777-4 2004 Among these proteins, the expression levels of p21 and pRB showed significant differences that were associated with the IC50 values for CPT-11 (P = 0.0339 and P = 0.0109, respectively). Irinotecan 136-142 RB transcriptional corepressor 1 Homo sapiens 55-58 15100172-0 2004 Hydrolysis of irinotecan and its oxidative metabolites, 7-ethyl-10-[4-N-(5-aminopentanoic acid)-1-piperidino] carbonyloxycamptothecin and 7-ethyl-10-[4-(1-piperidino)-1-amino]-carbonyloxycamptothecin, by human carboxylesterases CES1A1, CES2, and a newly expressed carboxylesterase isoenzyme, CES3. Irinotecan 14-24 carboxylesterase 2 Homo sapiens 210-227 15100172-0 2004 Hydrolysis of irinotecan and its oxidative metabolites, 7-ethyl-10-[4-N-(5-aminopentanoic acid)-1-piperidino] carbonyloxycamptothecin and 7-ethyl-10-[4-(1-piperidino)-1-amino]-carbonyloxycamptothecin, by human carboxylesterases CES1A1, CES2, and a newly expressed carboxylesterase isoenzyme, CES3. Irinotecan 14-24 carboxylesterase 2 Homo sapiens 236-240 15132777-5 2004 Namely, the expression of p21 or pRB independently could be a good indicator of CPT-11 efficacy in ESCC. Irinotecan 80-86 H3 histone pseudogene 16 Homo sapiens 26-29 15132777-5 2004 Namely, the expression of p21 or pRB independently could be a good indicator of CPT-11 efficacy in ESCC. Irinotecan 80-86 RB transcriptional corepressor 1 Homo sapiens 33-36 15100172-0 2004 Hydrolysis of irinotecan and its oxidative metabolites, 7-ethyl-10-[4-N-(5-aminopentanoic acid)-1-piperidino] carbonyloxycamptothecin and 7-ethyl-10-[4-(1-piperidino)-1-amino]-carbonyloxycamptothecin, by human carboxylesterases CES1A1, CES2, and a newly expressed carboxylesterase isoenzyme, CES3. Irinotecan 14-24 carboxylesterase 3 Homo sapiens 292-296 15143945-0 2004 Effect of human noxa on irinotecan-induced apoptosis in human gastric carcinoma cell lines. Irinotecan 24-34 phorbol-12-myristate-13-acetate-induced protein 1 Homo sapiens 16-20 15100172-6 2004 Carboxylesterases metabolize the carbamate prodrug 7-ethyl-10-[4-(1-piperidino)-1-piperidino] carbonyloxycamptothecin (CPT-11; irinotecan) to its active metabolite 7-ethyl-10-hydroxycamptothecin (SN-38), a potent topoisomerase I inhibitor. Irinotecan 51-117 carboxylesterase 2 Homo sapiens 0-17 15100172-6 2004 Carboxylesterases metabolize the carbamate prodrug 7-ethyl-10-[4-(1-piperidino)-1-piperidino] carbonyloxycamptothecin (CPT-11; irinotecan) to its active metabolite 7-ethyl-10-hydroxycamptothecin (SN-38), a potent topoisomerase I inhibitor. Irinotecan 127-137 carboxylesterase 2 Homo sapiens 0-17 15100172-6 2004 Carboxylesterases metabolize the carbamate prodrug 7-ethyl-10-[4-(1-piperidino)-1-piperidino] carbonyloxycamptothecin (CPT-11; irinotecan) to its active metabolite 7-ethyl-10-hydroxycamptothecin (SN-38), a potent topoisomerase I inhibitor. Irinotecan 164-194 carboxylesterase 2 Homo sapiens 0-17 15100172-6 2004 Carboxylesterases metabolize the carbamate prodrug 7-ethyl-10-[4-(1-piperidino)-1-piperidino] carbonyloxycamptothecin (CPT-11; irinotecan) to its active metabolite 7-ethyl-10-hydroxycamptothecin (SN-38), a potent topoisomerase I inhibitor. Irinotecan 196-201 carboxylesterase 2 Homo sapiens 0-17 15100172-8 2004 In this study, we investigate whether these oxidative metabolites, NPC and APC, can be metabolized to SN-38 by purified human carboxylesterases, CES1A1, CES2, and CES3. Irinotecan 102-107 carboxylesterase 2 Homo sapiens 126-143 15100172-8 2004 In this study, we investigate whether these oxidative metabolites, NPC and APC, can be metabolized to SN-38 by purified human carboxylesterases, CES1A1, CES2, and CES3. Irinotecan 102-107 carboxylesterase 2 Homo sapiens 153-157 15100172-8 2004 In this study, we investigate whether these oxidative metabolites, NPC and APC, can be metabolized to SN-38 by purified human carboxylesterases, CES1A1, CES2, and CES3. Irinotecan 102-107 carboxylesterase 3 Homo sapiens 163-167 15100172-9 2004 We find that CPT-11, APC, and NPC can all be metabolized by carboxylesterases to SN-38. Irinotecan 81-86 carboxylesterase 2 Homo sapiens 60-77 15143945-6 2004 MKN45 that has wild-type p53 showed severe inhibition by irinotecan compared with MKN28, which has mutated p53. Irinotecan 57-67 tumor protein p53 Homo sapiens 25-28 15255290-0 2004 Transport of SN-38 by the wild type of human ABC transporter ABCG2 and its inhibition by quercetin, a natural flavonoid. Irinotecan 13-18 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 61-66 15007088-0 2004 Genetic variants in the UDP-glucuronosyltransferase 1A1 gene predict the risk of severe neutropenia of irinotecan. Irinotecan 103-113 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 24-55 15007088-2 2004 UDP-glucuronosyltransferase 1A1 (UGT1A1) catalyzes the glucuronidation of the active metabolite SN-38. Irinotecan 96-101 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-31 15007088-2 2004 UDP-glucuronosyltransferase 1A1 (UGT1A1) catalyzes the glucuronidation of the active metabolite SN-38. Irinotecan 96-101 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-39 15007088-14 2004 CONCLUSION: UGT1A1 genotype and total bilirubin levels are strongly associated with severe neutropenia, and could be used to identify cancer patients predisposed to the severe toxicity of irinotecan. Irinotecan 188-198 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-18 15059881-3 2004 Previously, 4-anilinoquinazoline TKIs have been shown to inhibit the function of the breast cancer resistance-associated drug transporter (ABCG2), reversing resistance to camptothecin derivatives topotecan and SN-38. Irinotecan 210-215 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 139-144 15059881-5 2004 Here, we show that imatinib mesylate potently reverses ABCG2-mediated resistance to topotecan and SN-38 and significantly increases accumulation of topotecan only in cells expressing functional ABCG2. Irinotecan 98-103 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 55-60 15059881-0 2004 Imatinib mesylate is a potent inhibitor of the ABCG2 (BCRP) transporter and reverses resistance to topotecan and SN-38 in vitro. Irinotecan 113-118 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 47-52 15059881-0 2004 Imatinib mesylate is a potent inhibitor of the ABCG2 (BCRP) transporter and reverses resistance to topotecan and SN-38 in vitro. Irinotecan 113-118 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 54-58 15255290-1 2004 Irinotecan (CPT-11) is a widely-used potent anticancer drug that inhibits mammalian DNA topoisomerase I, however overexpression of the ATP-binding cassette transporter ABCG2 can confer cancer cells resistance to SN-38, the active form of CPT-11. Irinotecan 0-10 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 168-173 15255290-1 2004 Irinotecan (CPT-11) is a widely-used potent anticancer drug that inhibits mammalian DNA topoisomerase I, however overexpression of the ATP-binding cassette transporter ABCG2 can confer cancer cells resistance to SN-38, the active form of CPT-11. Irinotecan 12-18 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 168-173 15255290-1 2004 Irinotecan (CPT-11) is a widely-used potent anticancer drug that inhibits mammalian DNA topoisomerase I, however overexpression of the ATP-binding cassette transporter ABCG2 can confer cancer cells resistance to SN-38, the active form of CPT-11. Irinotecan 212-217 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 168-173 15255290-1 2004 Irinotecan (CPT-11) is a widely-used potent anticancer drug that inhibits mammalian DNA topoisomerase I, however overexpression of the ATP-binding cassette transporter ABCG2 can confer cancer cells resistance to SN-38, the active form of CPT-11. Irinotecan 238-244 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 168-173 15255290-2 2004 In the present study, we have examined the contribution of three variant forms of ABCG2 to SN-38 resistance. Irinotecan 91-96 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 82-87 15255290-3 2004 Exogenous expression of the Arg482 (wild type), Gly482, and Thr482 variants of ABCG2 conferred HEK293 cells resistance to SN-38 by 15.0-, 5.0-, and 5.3-fold, respectively. Irinotecan 122-127 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 79-84 15255290-4 2004 In plasma membrane vesicles prepared from the ABCG2 variant cDNA-transfected HEK293 cells, [Arg482]ABCG2 transported SN-38 and its glucuronide conjugate in an ATP-dependent manner; however, only minimal transport activities were observed with the other variants (Gly482 and Thr482). Irinotecan 117-122 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 46-51 15255290-4 2004 In plasma membrane vesicles prepared from the ABCG2 variant cDNA-transfected HEK293 cells, [Arg482]ABCG2 transported SN-38 and its glucuronide conjugate in an ATP-dependent manner; however, only minimal transport activities were observed with the other variants (Gly482 and Thr482). Irinotecan 117-122 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 99-104 15255290-7 2004 When [Arg482]ABCG2-transfected HEK293 cells were incubated with SN-38 in the presence of 20 microM quercetin, cellular resistance to SN-38 was partly reversed. Irinotecan 64-69 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 13-18 15255290-7 2004 When [Arg482]ABCG2-transfected HEK293 cells were incubated with SN-38 in the presence of 20 microM quercetin, cellular resistance to SN-38 was partly reversed. Irinotecan 133-138 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 13-18 15001986-6 2004 Activated wt-p53 increased apoptosis and enhanced sensitivity to CPT-11. Irinotecan 65-71 tumor protein p53 Homo sapiens 13-16 15134628-5 2004 CPT-11 was administered on days 1, 8, 15, and 22 at 225 mg/m2 for patients receiving CYP3A1- or CYP3A4-inducing anticonvulsants and at 125 mg/m2 for those not on these medications. Irinotecan 0-6 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 96-102 15041737-0 2004 Characterization of p53 wild-type and null isogenic colorectal cancer cell lines resistant to 5-fluorouracil, oxaliplatin, and irinotecan. Irinotecan 127-137 tumor protein p53 Homo sapiens 20-23 15001986-0 2004 Enhanced sensitivity to irinotecan by Cdk1 inhibition in the p53-deficient HT29 human colon cancer cell line. Irinotecan 24-34 cyclin dependent kinase 1 Homo sapiens 38-42 15001986-0 2004 Enhanced sensitivity to irinotecan by Cdk1 inhibition in the p53-deficient HT29 human colon cancer cell line. Irinotecan 24-34 tumor protein p53 Homo sapiens 61-64 15001986-9 2004 Cdk1 induction was exploited in vivo to improve the sensitivity to CPT-11 by additional treatment with the cdk-I CYC-202. Irinotecan 67-73 cyclin dependent kinase 1 Homo sapiens 0-4 15001986-2 2004 Irinotecan (CPT-11), a DNA topoisomerase 1 inhibitor, has been recently incorporated to the adjuvant therapy. Irinotecan 0-10 DNA topoisomerase I Homo sapiens 23-42 15001986-2 2004 Irinotecan (CPT-11), a DNA topoisomerase 1 inhibitor, has been recently incorporated to the adjuvant therapy. Irinotecan 12-18 DNA topoisomerase I Homo sapiens 23-42 15001986-3 2004 Since the DNA-damage checkpoint depends on p53 activation, the status of p53 might critically influence the response to CPT-11. Irinotecan 120-126 tumor protein p53 Homo sapiens 43-46 15001986-10 2004 We demonstrate a gain of sensitivity to CPT-11 in a p53-mutated colon cancer model either by restoring wild-type p53 function or by sequential treatment with cdk-Is. Irinotecan 40-46 tumor protein p53 Homo sapiens 52-55 15001986-3 2004 Since the DNA-damage checkpoint depends on p53 activation, the status of p53 might critically influence the response to CPT-11. Irinotecan 120-126 tumor protein p53 Homo sapiens 73-76 15001986-10 2004 We demonstrate a gain of sensitivity to CPT-11 in a p53-mutated colon cancer model either by restoring wild-type p53 function or by sequential treatment with cdk-Is. Irinotecan 40-46 tumor protein p53 Homo sapiens 113-116 14618629-1 2004 Breast cancer resistance protein (BCRP/ABCG2) of an ATP-binding cassette half-transporter confers resistance against mitoxantrone and camptothecin derivatives of topotecan and irinotecan. Irinotecan 176-186 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 0-32 14973080-9 2004 In 4-day cytotoxicity assays, ABCG2-mediated resistance to SN-38 and topotecan was abrogated in ABCG2-transfected HEK-293 cells treated with 1 micro M tariquidar, and ABCG2-transfected cells were 6-7-fold resistant to UCN-01. Irinotecan 59-64 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 30-35 14973080-9 2004 In 4-day cytotoxicity assays, ABCG2-mediated resistance to SN-38 and topotecan was abrogated in ABCG2-transfected HEK-293 cells treated with 1 micro M tariquidar, and ABCG2-transfected cells were 6-7-fold resistant to UCN-01. Irinotecan 59-64 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 96-101 14973080-9 2004 In 4-day cytotoxicity assays, ABCG2-mediated resistance to SN-38 and topotecan was abrogated in ABCG2-transfected HEK-293 cells treated with 1 micro M tariquidar, and ABCG2-transfected cells were 6-7-fold resistant to UCN-01. Irinotecan 59-64 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 96-101 14973083-1 2004 The breast cancer resistance protein (BCRP) is an ATP-binding cassette half transporter that confers resistance to anticancer drugs such as mitoxantrone, anthracyclines, topotecan, and SN-38. Irinotecan 185-190 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 4-36 14973083-1 2004 The breast cancer resistance protein (BCRP) is an ATP-binding cassette half transporter that confers resistance to anticancer drugs such as mitoxantrone, anthracyclines, topotecan, and SN-38. Irinotecan 185-190 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 38-42 14973083-1 2004 The breast cancer resistance protein (BCRP) is an ATP-binding cassette half transporter that confers resistance to anticancer drugs such as mitoxantrone, anthracyclines, topotecan, and SN-38. Irinotecan 185-190 ATP binding cassette subfamily B member 6 (Langereis blood group) Homo sapiens 50-87 15350151-5 2004 Drugs most prominently affected and contraindicated for concomitant use with St John"s wort are metabolised via both CYP3A4 and P-glycoprotein pathways, including HIV protease inhibitors, HIV non-nucleoside reverse transcriptase inhibitors (only CYP3A4), the immunosuppressants ciclosporin and tacrolimus, and the antineoplastic agents irinotecan and imatinib mesylate. Irinotecan 336-346 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 117-123 14658007-7 2004 As expected, in vitro studies revealed that a 5-day exposure to TSP-1 at concentrations up to 130 microg/ml was not cytotoxic alone and did not affect the cytotoxicity of CPT-11, or of its active metabolite SN38, in HT-29 cells. Irinotecan 207-211 thrombospondin 1 Homo sapiens 64-69 14962716-2 2004 In vitro studies have shown that irinotecan downregulates thymidylate synthase (TS) expression in tumour cells, leading to synergy between irinotecan and 5-FU that is maximal when irinotecan is given 24 h prior to 5-FU. Irinotecan 33-43 thymidylate synthetase Homo sapiens 58-78 14962716-2 2004 In vitro studies have shown that irinotecan downregulates thymidylate synthase (TS) expression in tumour cells, leading to synergy between irinotecan and 5-FU that is maximal when irinotecan is given 24 h prior to 5-FU. Irinotecan 139-149 thymidylate synthetase Homo sapiens 58-78 14962716-2 2004 In vitro studies have shown that irinotecan downregulates thymidylate synthase (TS) expression in tumour cells, leading to synergy between irinotecan and 5-FU that is maximal when irinotecan is given 24 h prior to 5-FU. Irinotecan 139-149 thymidylate synthetase Homo sapiens 58-78 15195891-7 2004 Favourable results were reported for the monoclonal antibody C225 (cetuximab) against the epidermal growth factor receptor in patients with ACC refractory to irinotecan. Irinotecan 158-168 epidermal growth factor receptor Homo sapiens 90-122 14967028-0 2004 Molecular modeling of CPT-11 metabolism by carboxylesterases (CEs): use of pnb CE as a model. Irinotecan 22-28 carboxylesterase 1 Homo sapiens 43-60 14967028-1 2004 CPT-11 is a prodrug that is converted in vivo to the topoisomerase I poison SN-38 by carboxylesterases (CEs). Irinotecan 0-6 carboxylesterase 1 Homo sapiens 85-102 14967028-1 2004 CPT-11 is a prodrug that is converted in vivo to the topoisomerase I poison SN-38 by carboxylesterases (CEs). Irinotecan 76-81 carboxylesterase 1 Homo sapiens 85-102 14967028-3 2004 Despite extensive sequence homology, however, the human homologues of this protein, hCE1 and hiCE, metabolize CPT-11 with significantly lower efficiencies. Irinotecan 110-116 carboxylesterase 1 Homo sapiens 84-88 14961077-4 2004 Using 19 glioblastoma cell lines, including 15 low-passage ex vivo cell lines derived from patients, as well as isogenic glioblastoma cells varying in p53 status, we show that clinically relevant levels of SN-38 potently induce cell cycle arrest and temporary senescence in glioblastoma cells with wild-type p53 while causing massive apoptosis in p53-deficient cells (P<0.0002). Irinotecan 206-211 tumor protein p53 Homo sapiens 151-154 14961077-4 2004 Using 19 glioblastoma cell lines, including 15 low-passage ex vivo cell lines derived from patients, as well as isogenic glioblastoma cells varying in p53 status, we show that clinically relevant levels of SN-38 potently induce cell cycle arrest and temporary senescence in glioblastoma cells with wild-type p53 while causing massive apoptosis in p53-deficient cells (P<0.0002). Irinotecan 206-211 tumor protein p53 Homo sapiens 308-311 14961077-4 2004 Using 19 glioblastoma cell lines, including 15 low-passage ex vivo cell lines derived from patients, as well as isogenic glioblastoma cells varying in p53 status, we show that clinically relevant levels of SN-38 potently induce cell cycle arrest and temporary senescence in glioblastoma cells with wild-type p53 while causing massive apoptosis in p53-deficient cells (P<0.0002). Irinotecan 206-211 tumor protein p53 Homo sapiens 308-311 15075664-10 2004 Furthermore, the combination of S-1 with cisplatin (CDDP), irinotecan or docetaxel for the treatment of gastric cancer and with CDDP for non-small cell and pancreatic cancer is feasible and active. Irinotecan 59-69 proteasome 26S subunit, non-ATPase 1 Homo sapiens 32-35 15641483-0 2004 Low overexpression of HER-2/neu in advanced colorectal cancer limits the usefulness of trastuzumab (Herceptin) and irinotecan as therapy. Irinotecan 115-125 erb-b2 receptor tyrosine kinase 2 Homo sapiens 22-31 15641483-2 2004 BACKGROUND: To determine the response rate of trastuzumab and irinotecan in HER-2/neu overexpressing advanced colorectal cancer (CRC), determine the frequency of HER-2/neu expression in CRC, and evaluate the pharmacokinetics of trastuzumab in a phase II study. Irinotecan 62-72 erb-b2 receptor tyrosine kinase 2 Homo sapiens 76-85 14618629-1 2004 Breast cancer resistance protein (BCRP/ABCG2) of an ATP-binding cassette half-transporter confers resistance against mitoxantrone and camptothecin derivatives of topotecan and irinotecan. Irinotecan 176-186 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 34-38 14618629-1 2004 Breast cancer resistance protein (BCRP/ABCG2) of an ATP-binding cassette half-transporter confers resistance against mitoxantrone and camptothecin derivatives of topotecan and irinotecan. Irinotecan 176-186 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 39-44 14618629-1 2004 Breast cancer resistance protein (BCRP/ABCG2) of an ATP-binding cassette half-transporter confers resistance against mitoxantrone and camptothecin derivatives of topotecan and irinotecan. Irinotecan 176-186 ATP binding cassette subfamily B member 6 (Langereis blood group) Homo sapiens 52-89 14618629-7 2004 Compared to the parental PC-6 cells, PC-6/SN2-5H2 cells were 141-, 173- and 57.2-fold resistant to topotecan, SN-38 and mitoxantrone, respectively. Irinotecan 110-115 proprotein convertase subtilisin/kexin type 5 Homo sapiens 37-41 14645705-2 2003 Mismatch-repair-deficient colonic tumors evade TRAIL-induced apoptosis through mutational inactivation of Bax, but chemotherapeutics including Camptosar (CPT-11) restore TRAIL sensitivity. Irinotecan 143-152 TNF superfamily member 10 Homo sapiens 170-175 15031593-9 2004 The monoclonal EGF receptor antibody cetuximab alone and in combination with irinotecan is active in second-line treatment. Irinotecan 77-87 epidermal growth factor Homo sapiens 15-18 15031593-10 2004 The VEGF antibody bevacizumab prolongs survival when given in combination with 5-FU/FA and irinotecan. Irinotecan 91-101 vascular endothelial growth factor A Homo sapiens 4-8 15655572-9 2004 The anti-VEGF bevacizumab increases the efficacy of first-line irinotecan therapy, while the addition of cetuximab restores irinotecan sensitivity in second line treatment of stage IV CRC. Irinotecan 63-73 vascular endothelial growth factor A Homo sapiens 9-13 14566825-2 2003 Expression of wild-type BCRP confers resistance to multiple chemotherapeutic agents such as mitoxantrone, SN-38 and topotecan, but not to doxorubicin. Irinotecan 106-111 ATP binding cassette subfamily G member 2 (Junior blood group) Mus musculus 24-28 14566825-6 2003 Cells transfected with N557D-BCRP cDNA showed similar resistance to mitoxantrone but lower resistance to SN-38 than the wild-type BCRP-transfected cells. Irinotecan 105-110 ATP binding cassette subfamily G member 2 (Junior blood group) Mus musculus 29-33 14566825-7 2003 Cells transfected with N557E-, H630E- or H630L-BCRP cDNA showed similar degrees of resistance to mitoxantrone and SN-38. Irinotecan 114-119 ATP binding cassette subfamily G member 2 (Junior blood group) Mus musculus 47-51 14645705-2 2003 Mismatch-repair-deficient colonic tumors evade TRAIL-induced apoptosis through mutational inactivation of Bax, but chemotherapeutics including Camptosar (CPT-11) restore TRAIL sensitivity. Irinotecan 154-160 TNF superfamily member 10 Homo sapiens 170-175 14645705-4 2003 Here, we imaged p53 transcriptional activity in Bax-/- carcinomas by using bioluminescence, in vivo, and find that p53 is required for sensitization to TRAIL by CPT-11. Irinotecan 161-167 BCL2 associated X, apoptosis regulator Homo sapiens 48-51 14645705-4 2003 Here, we imaged p53 transcriptional activity in Bax-/- carcinomas by using bioluminescence, in vivo, and find that p53 is required for sensitization to TRAIL by CPT-11. Irinotecan 161-167 tumor protein p53 Homo sapiens 115-118 14645705-4 2003 Here, we imaged p53 transcriptional activity in Bax-/- carcinomas by using bioluminescence, in vivo, and find that p53 is required for sensitization to TRAIL by CPT-11. Irinotecan 161-167 TNF superfamily member 10 Homo sapiens 152-157 14645705-6 2003 Caspase 8 inhibition protects both CPT-11 pretreated wild-type and Bax-/- HCT116 cells from TRAIL-induced apoptosis, whereas caspase 9 inhibition only rescued the wild-type HCT116 cells from death induced by TRAIL. Irinotecan 35-41 caspase 8 Homo sapiens 0-9 14645705-6 2003 Caspase 8 inhibition protects both CPT-11 pretreated wild-type and Bax-/- HCT116 cells from TRAIL-induced apoptosis, whereas caspase 9 inhibition only rescued the wild-type HCT116 cells from death induced by TRAIL. Irinotecan 35-41 TNF superfamily member 10 Homo sapiens 92-97 14645705-8 2003 In contrast to Bax-/- cells, Bak-deficient human cancers undergo apoptosis in response to TRAIL or CPT-11, implying that these proteins have nonoverlapping functions. Irinotecan 99-105 BCL2 antagonist/killer 1 Homo sapiens 29-32 14679008-3 2003 Identification of the UGT responsible for glucuronidation of SN-38 and the anthraquinone NU/ICRF 505 was achieved by first using a panel of human cDNA-expressed isozymes to measure conjugating activity. Irinotecan 61-66 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 22-25 14617791-1 2003 7-Ethyl-10-[4-(1-piperidino)-1-piperidino]carbonyloxycamptothecin (irinotecan, CPT-11) is a camptothecin prodrug that is metabolized by carboxylesterases (CE) to the active metabolite 7-ethyl-10-hydroxycamptothecin (SN-38), a topoisomerase I inhibitor. Irinotecan 0-65 carboxylesterase 1 Homo sapiens 136-153 14716146-4 2003 The monoclonal antibody cetuximab, targeted against EGFR, offers another new and very effective therapeutic option to patients with advanced irinotecan-refractory colorectal cancer--even those who are already pretreated with oxaliplatin. Irinotecan 141-151 epidermal growth factor receptor Homo sapiens 52-56 14612912-4 2003 In cytotoxicity assays, all ABCG2 proteins conferred high levels of resistance to mitoxantrone, SN-38, and topotecan, while mutant ABCG2 also exhibited a gain of function for mitoxantrone as they conferred a four-fold greater resistance compared to wild type. Irinotecan 96-101 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 28-33 14646693-0 2003 Haplotype analysis of ABCB1/MDR1 blocks in a Japanese population reveals genotype-dependent renal clearance of irinotecan. Irinotecan 111-121 ATP binding cassette subfamily B member 1 Homo sapiens 22-27 14646693-0 2003 Haplotype analysis of ABCB1/MDR1 blocks in a Japanese population reveals genotype-dependent renal clearance of irinotecan. Irinotecan 111-121 ATP binding cassette subfamily B member 1 Homo sapiens 28-32 14763144-10 2003 Partial responses were observed with cetuximab either alone (RR: 10%) or in combination with CPT-11 (RR: 22%) in patients with CPT-11 refractory advanced CRC which expressed EGF-R. Irinotecan 93-99 epidermal growth factor receptor Homo sapiens 174-179 14763144-10 2003 Partial responses were observed with cetuximab either alone (RR: 10%) or in combination with CPT-11 (RR: 22%) in patients with CPT-11 refractory advanced CRC which expressed EGF-R. Irinotecan 127-133 epidermal growth factor receptor Homo sapiens 174-179 14586211-0 2003 Weekly regimen of irinotecan/docetaxel in previously treated non-small cell lung cancer patients and correlation with uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) polymorphism. Irinotecan 18-28 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 118-165 14586211-0 2003 Weekly regimen of irinotecan/docetaxel in previously treated non-small cell lung cancer patients and correlation with uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) polymorphism. Irinotecan 18-28 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 167-173 14617791-1 2003 7-Ethyl-10-[4-(1-piperidino)-1-piperidino]carbonyloxycamptothecin (irinotecan, CPT-11) is a camptothecin prodrug that is metabolized by carboxylesterases (CE) to the active metabolite 7-ethyl-10-hydroxycamptothecin (SN-38), a topoisomerase I inhibitor. Irinotecan 67-77 carboxylesterase 1 Homo sapiens 136-153 14617791-1 2003 7-Ethyl-10-[4-(1-piperidino)-1-piperidino]carbonyloxycamptothecin (irinotecan, CPT-11) is a camptothecin prodrug that is metabolized by carboxylesterases (CE) to the active metabolite 7-ethyl-10-hydroxycamptothecin (SN-38), a topoisomerase I inhibitor. Irinotecan 79-85 carboxylesterase 1 Homo sapiens 136-153 14617791-1 2003 7-Ethyl-10-[4-(1-piperidino)-1-piperidino]carbonyloxycamptothecin (irinotecan, CPT-11) is a camptothecin prodrug that is metabolized by carboxylesterases (CE) to the active metabolite 7-ethyl-10-hydroxycamptothecin (SN-38), a topoisomerase I inhibitor. Irinotecan 184-214 carboxylesterase 1 Homo sapiens 136-153 14617791-1 2003 7-Ethyl-10-[4-(1-piperidino)-1-piperidino]carbonyloxycamptothecin (irinotecan, CPT-11) is a camptothecin prodrug that is metabolized by carboxylesterases (CE) to the active metabolite 7-ethyl-10-hydroxycamptothecin (SN-38), a topoisomerase I inhibitor. Irinotecan 216-221 carboxylesterase 1 Homo sapiens 136-153 14584274-7 2003 The circulating TPO concentration remained almost constant in the irinotecan group, with no significant increases, while that in the docetaxel group was increased significantly on day 4 (p < 0.05), day 8 (p < 0.05), and day 15 (p < 0.01) after therapy. Irinotecan 66-76 thrombopoietin Homo sapiens 16-19 14534729-0 2003 The relative contributions of carboxylesterase and beta-glucuronidase in the formation of SN-38 in human colorectal tumours. Irinotecan 90-95 glucuronidase beta Homo sapiens 51-69 14534729-3 2003 SN-38 is subsequently metabolised to its inactive glucuronide, SN-38G, which can however be reactivated to SN-38 by beta-glucuronidase. Irinotecan 0-5 glucuronidase beta Homo sapiens 116-134 14534729-3 2003 SN-38 is subsequently metabolised to its inactive glucuronide, SN-38G, which can however be reactivated to SN-38 by beta-glucuronidase. Irinotecan 63-68 glucuronidase beta Homo sapiens 116-134 14534729-3 2003 SN-38 is subsequently metabolised to its inactive glucuronide, SN-38G, which can however be reactivated to SN-38 by beta-glucuronidase. Irinotecan 63-68 glucuronidase beta Homo sapiens 116-134 14534729-4 2003 The purpose of this study was to examine the role of carboxylesterases and beta-glucuronidase in the in vitro production of SN-38 in human colorectal tumours. Irinotecan 124-129 glucuronidase beta Homo sapiens 75-93 14534729-8 2003 Therefore, tumour beta-glucuronidase may play a significant role in the exposure of tumours to SN-38 in vivo, particularly during prolonged infusions of CPT-11. Irinotecan 95-100 glucuronidase beta Homo sapiens 18-36 14534729-8 2003 Therefore, tumour beta-glucuronidase may play a significant role in the exposure of tumours to SN-38 in vivo, particularly during prolonged infusions of CPT-11. Irinotecan 153-159 glucuronidase beta Homo sapiens 18-36 14522368-7 2003 Cytochrome P450 3A4 inducing anti-epileptic drugs like phenytoin, carbamazepine and phenobarbital may significantly increase the metabolism of many chemotherapeutic agents like CPT11 and paclitaxel (but also of newer biological agents like many tyrosine kinase inhibitors). Irinotecan 177-182 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 0-19 14581373-9 2003 Only CES2 gene expression correlated with the carboxylesterase activity assays (P < 0.01) with CPT-11 and 4-methylumbelliferyl acetate as substrates. Irinotecan 98-104 carboxylesterase 2 Homo sapiens 5-9 14581373-12 2003 CONCLUSIONS: We conclude that CES2 is the most abundant carboxylesterase in colon tumors that is responsible for CPT-11 hydrolysis. Irinotecan 113-119 carboxylesterase 2 Homo sapiens 30-34 14581373-13 2003 This pilot study reinforces the hypothesis that there is a large interindividual variation in expression of carboxylesterases that may contribute to variation in therapeutic outcome and/or toxicity of CPT-11 therapy for colon cancer. Irinotecan 201-207 carboxylesterase 2 Homo sapiens 108-125 14584274-8 2003 There was also a significant difference between the docetaxel group and irinotecan group with respect to circulating TPO concentration (p < 0.05). Irinotecan 72-82 thrombopoietin Homo sapiens 117-120 12911669-12 2003 CONCLUSIONS: We conclude that irinotecan causes diarrhea by inducing apoptosis and hypoproliferation in both the small and large intestines, and causes colonic damage with changes in goblet cells and mucin secretion. Irinotecan 30-40 solute carrier family 13 member 2 Rattus norvegicus 200-205 14522894-6 2003 A better response to irinotecan was observed in the patients whose tumors have lost BAX expression (P < 0.001). Irinotecan 21-31 BCL2 associated X, apoptosis regulator Homo sapiens 84-87 12912956-1 2003 PURPOSE: Breast cancer resistance protein (BCRP/ABCG2), an ATP binding cassette half-transporter, confers resistance to mitoxantrone, doxorubicin, and topoisomerase I inhibitors of irinotecan and topotecan. Irinotecan 181-191 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 9-41 12920197-1 2003 Breast cancer resistance protein (BCRP), an ATP-binding cassette transporter, confers resistance to a series of anticancer reagents such as mitoxantrone, 7-ethyl-10-hydroxycamptothecin, and topotecan. Irinotecan 154-184 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 0-32 12920197-1 2003 Breast cancer resistance protein (BCRP), an ATP-binding cassette transporter, confers resistance to a series of anticancer reagents such as mitoxantrone, 7-ethyl-10-hydroxycamptothecin, and topotecan. Irinotecan 154-184 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 34-38 12932648-5 2003 The SN-38 formulations were stable in human serum albumin and high lactone concentrations were observed even after 3 h. In vivo studies in nude mice showed prolonged half-life of the active (lactone form) drug in whole blood and increased efficacy compared to Camptosar in a mouse xenograft tumor model. Irinotecan 4-9 albumin Mus musculus 50-57 12960109-5 2003 The homozygous T allele of the ABCB1 1236C>T polymorphism was associated with significantly increased exposure to irinotecan (P = 0.038) and its active metabolite SN-38 (P = 0.031). Irinotecan 117-127 ATP binding cassette subfamily B member 1 Homo sapiens 31-36 12960109-5 2003 The homozygous T allele of the ABCB1 1236C>T polymorphism was associated with significantly increased exposure to irinotecan (P = 0.038) and its active metabolite SN-38 (P = 0.031). Irinotecan 166-171 ATP binding cassette subfamily B member 1 Homo sapiens 31-36 12960109-7 2003 The extent of SN-38 glucuronidation was slightly impaired in homozygous variants of UGT1A1*28, although differences were not statistically significant (P = 0.22). Irinotecan 14-19 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 84-90 12960109-8 2003 CONCLUSIONS: It is concluded that genotyping for ABCB1 1236C>T may be one of the factors assisting with dose optimization of irinotecan chemotherapy in cancer patients. Irinotecan 128-138 ATP binding cassette subfamily B member 1 Homo sapiens 49-54 12907654-3 2003 When combined with the topoisomerase I inhibitor CPT-11 (irinotecan), Apo2L/TRAIL exhibits enhanced apoptotic activity in C4-2 cells cultured in vitro as well as xenografted as tumors in vivo. Irinotecan 49-55 TNF superfamily member 10 Homo sapiens 70-75 12907654-3 2003 When combined with the topoisomerase I inhibitor CPT-11 (irinotecan), Apo2L/TRAIL exhibits enhanced apoptotic activity in C4-2 cells cultured in vitro as well as xenografted as tumors in vivo. Irinotecan 49-55 TNF superfamily member 10 Homo sapiens 76-81 12907654-3 2003 When combined with the topoisomerase I inhibitor CPT-11 (irinotecan), Apo2L/TRAIL exhibits enhanced apoptotic activity in C4-2 cells cultured in vitro as well as xenografted as tumors in vivo. Irinotecan 57-67 TNF superfamily member 10 Homo sapiens 70-75 12907654-3 2003 When combined with the topoisomerase I inhibitor CPT-11 (irinotecan), Apo2L/TRAIL exhibits enhanced apoptotic activity in C4-2 cells cultured in vitro as well as xenografted as tumors in vivo. Irinotecan 57-67 TNF superfamily member 10 Homo sapiens 76-81 12907654-10 2003 Activation of multiple caspases and PARP cleavage were also observed in the C4-2 tumors treated with doses resulting in effective tumor control at 42 days after Apo2L/TRAIL plus CPT-11 treatment. Irinotecan 178-184 caspase 8 Homo sapiens 23-31 12907654-10 2003 Activation of multiple caspases and PARP cleavage were also observed in the C4-2 tumors treated with doses resulting in effective tumor control at 42 days after Apo2L/TRAIL plus CPT-11 treatment. Irinotecan 178-184 poly(ADP-ribose) polymerase 1 Homo sapiens 36-40 12907654-13 2003 Our data indicate that the combined treatment of Apo2L/TRAIL and CPT-11 achieves tumor control in prostate cancer tumors through regulation of Bcl-2 family proteins and potent activation of caspases. Irinotecan 65-71 BCL2 apoptosis regulator Homo sapiens 143-148 12907654-13 2003 Our data indicate that the combined treatment of Apo2L/TRAIL and CPT-11 achieves tumor control in prostate cancer tumors through regulation of Bcl-2 family proteins and potent activation of caspases. Irinotecan 65-71 caspase 8 Homo sapiens 190-198 12914384-8 2003 UGT1A1 (uridine diphoshate-glucuronosyltransferase 1A1) is involved on irinotecan metabolism. Irinotecan 71-81 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 12914384-8 2003 UGT1A1 (uridine diphoshate-glucuronosyltransferase 1A1) is involved on irinotecan metabolism. Irinotecan 71-81 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 8-54 12914384-9 2003 MRP2 (multi-drug resistance associated protein) and MDR1 (multi-drug resistance gene) are involved in irinotecan as well as anthracyclines transport. Irinotecan 102-112 ATP binding cassette subfamily C member 2 Homo sapiens 0-4 12914384-9 2003 MRP2 (multi-drug resistance associated protein) and MDR1 (multi-drug resistance gene) are involved in irinotecan as well as anthracyclines transport. Irinotecan 102-112 ATP binding cassette subfamily B member 1 Homo sapiens 52-56 12912956-1 2003 PURPOSE: Breast cancer resistance protein (BCRP/ABCG2), an ATP binding cassette half-transporter, confers resistance to mitoxantrone, doxorubicin, and topoisomerase I inhibitors of irinotecan and topotecan. Irinotecan 181-191 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 43-47 12912956-1 2003 PURPOSE: Breast cancer resistance protein (BCRP/ABCG2), an ATP binding cassette half-transporter, confers resistance to mitoxantrone, doxorubicin, and topoisomerase I inhibitors of irinotecan and topotecan. Irinotecan 181-191 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 48-53 12912956-2 2003 Recently, we reported that BCRP efficiently transported SN-38 (the active metabolite of irinotecan) with a high affinity in lung cancer cells in vitro (K. Nakatomi et al., Biochem. Irinotecan 56-61 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 27-31 12912956-2 2003 Recently, we reported that BCRP efficiently transported SN-38 (the active metabolite of irinotecan) with a high affinity in lung cancer cells in vitro (K. Nakatomi et al., Biochem. Irinotecan 88-98 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 27-31 12912956-9 2003 RESULTS: The levels of BCRP mRNA expression in the cell lines were significantly correlated with the BCRP function and the sensitivity to SN-38 and topotecan. Irinotecan 138-143 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 23-27 12869632-5 2003 The role of the ATP-dependent drug transporter ABCG2 in CPT-11 cytotoxicity is unclear because some ABCG2 mutants confer camptothecin resistance, whereas others do not. Irinotecan 56-62 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 47-52 12871782-1 2003 The anticancer agent irinotecan (CPT-11) is a prodrug converted to its active form, SN-38, by human carboxylesterase (hCE) and the SN-38 is further metabolized to its inactive form, SN-38G. Irinotecan 21-31 RNA guanylyltransferase and 5'-phosphatase Homo sapiens 118-121 12871782-1 2003 The anticancer agent irinotecan (CPT-11) is a prodrug converted to its active form, SN-38, by human carboxylesterase (hCE) and the SN-38 is further metabolized to its inactive form, SN-38G. Irinotecan 33-39 RNA guanylyltransferase and 5'-phosphatase Homo sapiens 118-121 12871782-1 2003 The anticancer agent irinotecan (CPT-11) is a prodrug converted to its active form, SN-38, by human carboxylesterase (hCE) and the SN-38 is further metabolized to its inactive form, SN-38G. Irinotecan 84-89 RNA guanylyltransferase and 5'-phosphatase Homo sapiens 118-121 12871782-1 2003 The anticancer agent irinotecan (CPT-11) is a prodrug converted to its active form, SN-38, by human carboxylesterase (hCE) and the SN-38 is further metabolized to its inactive form, SN-38G. Irinotecan 131-136 RNA guanylyltransferase and 5'-phosphatase Homo sapiens 118-121 12871782-1 2003 The anticancer agent irinotecan (CPT-11) is a prodrug converted to its active form, SN-38, by human carboxylesterase (hCE) and the SN-38 is further metabolized to its inactive form, SN-38G. Irinotecan 131-136 RNA guanylyltransferase and 5'-phosphatase Homo sapiens 118-121 12730278-2 2003 Recent studies have revealed that other UGT1A isoforms, UGT1A7 and UGT1A9, also participate in SN-38 glucuronidation. Irinotecan 95-100 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 40-45 12730278-2 2003 Recent studies have revealed that other UGT1A isoforms, UGT1A7 and UGT1A9, also participate in SN-38 glucuronidation. Irinotecan 95-100 UDP glucuronosyltransferase family 1 member A7 Homo sapiens 56-62 12730278-2 2003 Recent studies have revealed that other UGT1A isoforms, UGT1A7 and UGT1A9, also participate in SN-38 glucuronidation. Irinotecan 95-100 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 67-73 12730278-3 2003 Although several genetic polymorphisms are reported for UGT1A1 and UGT1A7 that affect the SN-38 glucuronidation activities, no such polymorphisms have been identified for UGT1A9. Irinotecan 90-95 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 56-62 12730278-3 2003 Although several genetic polymorphisms are reported for UGT1A1 and UGT1A7 that affect the SN-38 glucuronidation activities, no such polymorphisms have been identified for UGT1A9. Irinotecan 90-95 UDP glucuronosyltransferase family 1 member A7 Homo sapiens 67-73 12730278-4 2003 In the present study, UGT1A9 exon 1 and its flanking regions were sequenced from 61 Japanese cancer patients who were all treated with irinotecan. Irinotecan 135-145 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 22-28 12730278-8 2003 The SN-38 glucuronidation efficiency (normalized Vmax/Km) of D256N was less than 5% that of wild-type UGT1A9. Irinotecan 4-9 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 102-108 12730278-10 2003 These findings highlight the importance of further studies into the potential influence of UGT1A9 D256N variant to irinotecan metabolism in vivo. Irinotecan 115-125 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 91-97 12869632-7 2003 Our results indicate that the expression of ABCG2 protects cells from CPT-11 toxicity, even in the presence of high levels of a rabbit liver carboxylesterase (rCE), which can efficiently activate the drug. Irinotecan 70-76 ATP-binding cassette sub-family G member 2 Oryctolagus cuniculus 44-49 12939466-0 2003 Gene-directed enzyme prodrug therapy for osteosarcoma: sensitization to CPT-11 in vitro and in vivo by adenoviral delivery of a gene encoding secreted carboxylesterase-2. Irinotecan 72-78 carboxylesterase 2 Homo sapiens 151-169 12682043-4 2003 The ABCG2-mediated transport of [3H]E1S was potently inhibited by SN-38 and many sulfate conjugates but not by glucuronide and glutathione conjugates or other anionic compounds. Irinotecan 66-71 ATP binding cassette subfamily G member 2 (Junior blood group) Mus musculus 4-9 12939466-5 2003 GDEPT aims at high production of CE2 at the tumor site, resulting in efficient local conversion of CPT-11 into SN-38. Irinotecan 99-105 carboxylesterase 2 Homo sapiens 33-36 12939466-5 2003 GDEPT aims at high production of CE2 at the tumor site, resulting in efficient local conversion of CPT-11 into SN-38. Irinotecan 111-116 carboxylesterase 2 Homo sapiens 33-36 12855666-1 2003 PURPOSE: To identify determinants of the effect of antisense-mediated Bcl-xl down-regulation (Bcl-xl knockdown) on the response of colorectal cancer cells to SN38, the active metabolite of irinotecan, a topoisomerase I inhibitor licensed for colorectal cancer chemotherapy. Irinotecan 189-199 BCL2 like 1 Homo sapiens 70-76 12855666-1 2003 PURPOSE: To identify determinants of the effect of antisense-mediated Bcl-xl down-regulation (Bcl-xl knockdown) on the response of colorectal cancer cells to SN38, the active metabolite of irinotecan, a topoisomerase I inhibitor licensed for colorectal cancer chemotherapy. Irinotecan 189-199 BCL2 like 1 Homo sapiens 94-100 12886870-3 2003 Inhibition of the cyclooxygenase-2 (COX-2) enzyme, which is active in tumorigenesis, may augment efficacy and reduce toxicity of platinum/irinotecan combinations. Irinotecan 138-148 prostaglandin-endoperoxide synthase 2 Homo sapiens 18-34 12886870-3 2003 Inhibition of the cyclooxygenase-2 (COX-2) enzyme, which is active in tumorigenesis, may augment efficacy and reduce toxicity of platinum/irinotecan combinations. Irinotecan 138-148 prostaglandin-endoperoxide synthase 2 Homo sapiens 36-41 12817897-0 2003 Effect of P-glycoprotein modulator, cyclosporin A, on the gastrointestinal excretion of irinotecan and its metabolite SN-38 in rats. Irinotecan 88-98 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 10-24 12810652-8 2003 In addition, immunoblotting studies indicate that whereas increased BCRP expression is evident in cells selected for resistance to irinotecan, BCRP expression is not detectable in two different cell lines selected for resistance to 9-NC. Irinotecan 131-141 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 68-72 12817897-0 2003 Effect of P-glycoprotein modulator, cyclosporin A, on the gastrointestinal excretion of irinotecan and its metabolite SN-38 in rats. Irinotecan 118-123 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 10-24 12817897-1 2003 PURPOSE: The purpose of this work was to investigate the role of the hepatic and intestinal P-glycoprotein (P-gp) and canalicular multispecific organic anion transporter/multidrug resistance-associated protein 2 (cMOAT/MRP2) on both biliary excretion and intestinal exsorption of irinotecan hydrochloride (CPT-11) and its metabolite, SN-38, in the lactone and carboxylate forms. Irinotecan 280-304 ATP binding cassette subfamily C member 2 Rattus norvegicus 213-218 12817897-1 2003 PURPOSE: The purpose of this work was to investigate the role of the hepatic and intestinal P-glycoprotein (P-gp) and canalicular multispecific organic anion transporter/multidrug resistance-associated protein 2 (cMOAT/MRP2) on both biliary excretion and intestinal exsorption of irinotecan hydrochloride (CPT-11) and its metabolite, SN-38, in the lactone and carboxylate forms. Irinotecan 306-312 ATP binding cassette subfamily C member 2 Rattus norvegicus 213-218 12817897-1 2003 PURPOSE: The purpose of this work was to investigate the role of the hepatic and intestinal P-glycoprotein (P-gp) and canalicular multispecific organic anion transporter/multidrug resistance-associated protein 2 (cMOAT/MRP2) on both biliary excretion and intestinal exsorption of irinotecan hydrochloride (CPT-11) and its metabolite, SN-38, in the lactone and carboxylate forms. Irinotecan 334-339 ATP binding cassette subfamily C member 2 Rattus norvegicus 213-218 12817897-8 2003 CONCLUSIONS: The transports of CPT-11 and SN-38 via the biliary route seem to be essentially related with cMOAT/MRP2, whereas those of both compounds via the intestinal membrane seem to be related with P-gp. Irinotecan 31-37 ATP binding cassette subfamily C member 2 Rattus norvegicus 106-111 12817897-8 2003 CONCLUSIONS: The transports of CPT-11 and SN-38 via the biliary route seem to be essentially related with cMOAT/MRP2, whereas those of both compounds via the intestinal membrane seem to be related with P-gp. Irinotecan 31-37 ATP binding cassette subfamily C member 2 Rattus norvegicus 112-116 12817897-8 2003 CONCLUSIONS: The transports of CPT-11 and SN-38 via the biliary route seem to be essentially related with cMOAT/MRP2, whereas those of both compounds via the intestinal membrane seem to be related with P-gp. Irinotecan 31-37 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 202-206 12817897-8 2003 CONCLUSIONS: The transports of CPT-11 and SN-38 via the biliary route seem to be essentially related with cMOAT/MRP2, whereas those of both compounds via the intestinal membrane seem to be related with P-gp. Irinotecan 42-47 ATP binding cassette subfamily C member 2 Rattus norvegicus 106-111 12817897-8 2003 CONCLUSIONS: The transports of CPT-11 and SN-38 via the biliary route seem to be essentially related with cMOAT/MRP2, whereas those of both compounds via the intestinal membrane seem to be related with P-gp. Irinotecan 42-47 ATP binding cassette subfamily C member 2 Rattus norvegicus 112-116 12817897-8 2003 CONCLUSIONS: The transports of CPT-11 and SN-38 via the biliary route seem to be essentially related with cMOAT/MRP2, whereas those of both compounds via the intestinal membrane seem to be related with P-gp. Irinotecan 42-47 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 202-206 12712458-11 2003 The combination of a PKC inhibitor with CPT-11 or SN-38 led to decreased expression of the antiapoptotic protein bcl-2, and increased expression of the proapoptotic protein bax. Irinotecan 40-46 BCL2 apoptosis regulator Homo sapiens 113-118 12712458-11 2003 The combination of a PKC inhibitor with CPT-11 or SN-38 led to decreased expression of the antiapoptotic protein bcl-2, and increased expression of the proapoptotic protein bax. Irinotecan 40-46 BCL2 associated X, apoptosis regulator Homo sapiens 173-176 12712458-11 2003 The combination of a PKC inhibitor with CPT-11 or SN-38 led to decreased expression of the antiapoptotic protein bcl-2, and increased expression of the proapoptotic protein bax. Irinotecan 50-55 BCL2 apoptosis regulator Homo sapiens 113-118 12712458-11 2003 The combination of a PKC inhibitor with CPT-11 or SN-38 led to decreased expression of the antiapoptotic protein bcl-2, and increased expression of the proapoptotic protein bax. Irinotecan 50-55 BCL2 associated X, apoptosis regulator Homo sapiens 173-176 12847914-5 2003 Moreover, inhibition of FAK gene expression by a phosphorothioated antisense oligodeoxynucleotide targeting the portion of the gene encoding amino acids 262-268, increased the sensitivity of ZR-75-1, MDA-MB-231 and MCF7 breast cancer cells to treatment with TPT or CPT-11. Irinotecan 265-271 protein tyrosine kinase 2 Homo sapiens 24-27 12800608-6 2003 Two recent pharmacogenetic trials (one performed in the United States and the other in Japan) investigated the clinical significance of UGT1A1 gene mutations for both the pharmacokinetics of irinotecan metabolites and the toxicity profile. Irinotecan 191-201 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 136-142 12769780-8 2003 In particular, the UGT1A1*28 allele has been associated with an increased toxicity after irinotecan chemotherapy. Irinotecan 89-99 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 19-25 12800601-13 2003 COX-2 inhibitors also improve tumor response to chemotherapeutic agents, including irinotecan. Irinotecan 83-93 prostaglandin-endoperoxide synthase 2 Homo sapiens 0-5 12800601-14 2003 Additional therapeutic benefit was observed for celecoxib (Celebrex), a selective COX-2 inhibitor, consisting of a strong reduction in irinotecan-induced diarrhea. Irinotecan 135-145 prostaglandin-endoperoxide synthase 2 Homo sapiens 82-87 12800606-15 2003 The recommended phase II doses of irinotecan and temozolomide on treatment arms 1 and 3 remain to be determined as patient accrual is currently ongoing. Irinotecan 34-44 ADRM1 26S proteasome ubiquitin receptor Homo sapiens 75-87 12800608-0 2003 Irinotecan treatment in cancer patients with UGT1A1 polymorphisms. Irinotecan 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 45-51 12800608-1 2003 At the present time, pharmacogenetic investigation of irinotecan (CPT-11, Camptosar) therapy is mainly focused on the clinical relevance of genetic variation in the UDP-glucuronosyltransferase (UGT1A1) gene. Irinotecan 54-64 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 194-200 12800608-1 2003 At the present time, pharmacogenetic investigation of irinotecan (CPT-11, Camptosar) therapy is mainly focused on the clinical relevance of genetic variation in the UDP-glucuronosyltransferase (UGT1A1) gene. Irinotecan 66-72 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 194-200 12800608-1 2003 At the present time, pharmacogenetic investigation of irinotecan (CPT-11, Camptosar) therapy is mainly focused on the clinical relevance of genetic variation in the UDP-glucuronosyltransferase (UGT1A1) gene. Irinotecan 74-83 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 194-200 12800608-3 2003 UGT1A1 genotypes associated with Gilbert"s syndrome (a mild intermittent hyperbilirubinemia) are characterized by reduced glucuronidation of SN-38. Irinotecan 141-146 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 12704006-4 2003 The BCRP-overexpressing tumor cells are resistant to mitoxantrone, adriamycin, daunorubicin, etoposide, topotecan and irinotecan, but lack resistance to paclitaxel and vincristine. Irinotecan 118-128 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 4-8 12617927-2 2003 Since this prodrug is significantly less cytotoxic than SN-38 itself and efficiently releases the drug in vitro in the presence of beta-D-glucuronidase, it can be considered as an appropriate candidate for cancer treatment by a PMT strategy. Irinotecan 56-61 glucuronidase beta Homo sapiens 131-151 12485949-9 2003 M-LK transfected cells manifested more severe cytotoxicity than TGH transfected cells upon being exposed to irinotecan. Irinotecan 108-118 carboxylesterase 1D Mus musculus 64-67 12538824-0 2003 p53-mediated regulation of expression of a rabbit liver carboxylesterase confers sensitivity to 7-ethyl-10-[4-(1-piperidino)-1-piperidino]carbonyloxycamptothecin (CPT-11). Irinotecan 96-161 tumor protein p53 Homo sapiens 0-3 12538824-0 2003 p53-mediated regulation of expression of a rabbit liver carboxylesterase confers sensitivity to 7-ethyl-10-[4-(1-piperidino)-1-piperidino]carbonyloxycamptothecin (CPT-11). Irinotecan 96-161 liver carboxylesterase 1 Oryctolagus cuniculus 50-72 12538824-0 2003 p53-mediated regulation of expression of a rabbit liver carboxylesterase confers sensitivity to 7-ethyl-10-[4-(1-piperidino)-1-piperidino]carbonyloxycamptothecin (CPT-11). Irinotecan 163-169 tumor protein p53 Homo sapiens 0-3 12538824-0 2003 p53-mediated regulation of expression of a rabbit liver carboxylesterase confers sensitivity to 7-ethyl-10-[4-(1-piperidino)-1-piperidino]carbonyloxycamptothecin (CPT-11). Irinotecan 163-169 liver carboxylesterase 1 Oryctolagus cuniculus 50-72 12538824-3 2003 Upon transfection of these plasmids into cells expressing either wt or mutant p53, differential expression of the CE has been observed, resulting in sensitization of the cells expressing the latter protein to the anticancer prodrug irinotecan, 7-ethyl-10-[4-(1-piperidino)-1-piperidino] carb- onyloxycamptothecin (CPT-11). Irinotecan 232-242 tumor protein p53 Homo sapiens 78-81 12538824-3 2003 Upon transfection of these plasmids into cells expressing either wt or mutant p53, differential expression of the CE has been observed, resulting in sensitization of the cells expressing the latter protein to the anticancer prodrug irinotecan, 7-ethyl-10-[4-(1-piperidino)-1-piperidino] carb- onyloxycamptothecin (CPT-11). Irinotecan 244-312 tumor protein p53 Homo sapiens 78-81 12538824-3 2003 Upon transfection of these plasmids into cells expressing either wt or mutant p53, differential expression of the CE has been observed, resulting in sensitization of the cells expressing the latter protein to the anticancer prodrug irinotecan, 7-ethyl-10-[4-(1-piperidino)-1-piperidino] carb- onyloxycamptothecin (CPT-11). Irinotecan 314-320 tumor protein p53 Homo sapiens 78-81 12538824-5 2003 These studies indicate that the inactivation of wtp53 by mutant p53 in human tumor cells may be sufficient enough to generate a therapeutic window for enhanced cytotoxicity with CPT-11. Irinotecan 178-184 tumor protein p53 Homo sapiens 50-53 12569373-1 2003 To determine a standard combination chemotherapy for patients with advanced non-small-cell lung cancer (NSCLC), we conducted a phase III trial of irinotecan (CPT-11) to test the hypotheses that CPT-11+cisplatin is superior to cisplatin+vindesine and that CPT-11 monotherapy is not inferior to cisplatin+vindesine. Irinotecan 146-156 choline phosphotransferase 1 Homo sapiens 158-161 12570720-6 2003 Irinotecan is subjected to be shunted between CYP3A4 mediated oxidative metabolism to form two inactive metabolites APC or NPC and tissue carboxylesterase mediated hydrolysis to form SN-38 which is eventually detoxified via glucuronidation by UGT1A1 to form SN-38G. Irinotecan 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 46-52 12570720-6 2003 Irinotecan is subjected to be shunted between CYP3A4 mediated oxidative metabolism to form two inactive metabolites APC or NPC and tissue carboxylesterase mediated hydrolysis to form SN-38 which is eventually detoxified via glucuronidation by UGT1A1 to form SN-38G. Irinotecan 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 243-249 12570720-6 2003 Irinotecan is subjected to be shunted between CYP3A4 mediated oxidative metabolism to form two inactive metabolites APC or NPC and tissue carboxylesterase mediated hydrolysis to form SN-38 which is eventually detoxified via glucuronidation by UGT1A1 to form SN-38G. Irinotecan 183-188 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 243-249 12570720-6 2003 Irinotecan is subjected to be shunted between CYP3A4 mediated oxidative metabolism to form two inactive metabolites APC or NPC and tissue carboxylesterase mediated hydrolysis to form SN-38 which is eventually detoxified via glucuronidation by UGT1A1 to form SN-38G. Irinotecan 258-263 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 46-52 12570720-7 2003 The pharmacology of this compound is further complicated by the existence of genetic inter-individual differences in activation and deactivation enzymes of irinotecan (e.g., CYP3A4, CYP3A5, UGT1A1) and sharing competitive elimination pathways with many concomitant medications, such as anticonvulsants, St. John"s Wort, and ketoconazole. Irinotecan 156-166 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 174-180 12570720-7 2003 The pharmacology of this compound is further complicated by the existence of genetic inter-individual differences in activation and deactivation enzymes of irinotecan (e.g., CYP3A4, CYP3A5, UGT1A1) and sharing competitive elimination pathways with many concomitant medications, such as anticonvulsants, St. John"s Wort, and ketoconazole. Irinotecan 156-166 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 182-188 12570720-9 2003 This review highlights the latest findings in drug activation, transport mechanisms, glucuronidation, and CYP3A-mediated drug-drug interactions of irinotecan in order to unlock some of its complicated pharmacology and to provide ideas for relevant future studies into optimization of this promising agent. Irinotecan 147-157 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 106-111 15618737-1 2003 Eight novel single nucleotide polymorphisms (SNPs) were found in the gene encoding the ATP-binding cassette transporter, ABCG2/BCRP, from 60 Japanese individuals administered the anti-cancer drug irinotecan. Irinotecan 196-206 ATP binding cassette subfamily A member 4 Homo sapiens 87-119 12775012-11 2003 Several preliminary results of combination phase I/II studies of S-1 with CDDP or CPT-11 have recently been obtained, and phase II studies are in progress. Irinotecan 82-88 proteasome 26S subunit, non-ATPase 1 Homo sapiens 65-68 15618737-1 2003 Eight novel single nucleotide polymorphisms (SNPs) were found in the gene encoding the ATP-binding cassette transporter, ABCG2/BCRP, from 60 Japanese individuals administered the anti-cancer drug irinotecan. Irinotecan 196-206 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 121-126 15618737-1 2003 Eight novel single nucleotide polymorphisms (SNPs) were found in the gene encoding the ATP-binding cassette transporter, ABCG2/BCRP, from 60 Japanese individuals administered the anti-cancer drug irinotecan. Irinotecan 196-206 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 127-131 15618752-1 2003 Twelve novel single nucleotide polymorphisms (SNPs) were found in the CES2 gene from 153 Japanese individuals, who were administered irinotecan or steroidal drugs. Irinotecan 133-143 carboxylesterase 2 Homo sapiens 70-74 12607608-3 2002 Carboxylesterase-2 has been identified as the key enzyme in the metabolic activation of the irinotecan, a topoisomerase I inhibitor commonly used in the treatment of many solid tumors. Irinotecan 92-102 carboxylesterase 2 Homo sapiens 0-18 12485959-0 2003 Glucuronidation of 7-ethyl-10-hydroxycamptothecin (SN-38), an active metabolite of irinotecan (CPT-11), by human UGT1A1 variants, G71R, P229Q, and Y486D. Irinotecan 19-49 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 113-119 12485959-0 2003 Glucuronidation of 7-ethyl-10-hydroxycamptothecin (SN-38), an active metabolite of irinotecan (CPT-11), by human UGT1A1 variants, G71R, P229Q, and Y486D. Irinotecan 51-56 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 113-119 12485959-0 2003 Glucuronidation of 7-ethyl-10-hydroxycamptothecin (SN-38), an active metabolite of irinotecan (CPT-11), by human UGT1A1 variants, G71R, P229Q, and Y486D. Irinotecan 83-93 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 113-119 12485959-0 2003 Glucuronidation of 7-ethyl-10-hydroxycamptothecin (SN-38), an active metabolite of irinotecan (CPT-11), by human UGT1A1 variants, G71R, P229Q, and Y486D. Irinotecan 95-101 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 113-119 12485959-1 2003 7-Ethyl-10-hydroxycamptothecin (SN-38), an active metabolite of antitumor agent irinotecan (CPT-11), is conjugated and detoxified to SN-38-glucuronide by UDP-glucuronosyltransferase (UGT) 1A1. Irinotecan 0-30 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 154-191 12485959-1 2003 7-Ethyl-10-hydroxycamptothecin (SN-38), an active metabolite of antitumor agent irinotecan (CPT-11), is conjugated and detoxified to SN-38-glucuronide by UDP-glucuronosyltransferase (UGT) 1A1. Irinotecan 32-37 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 154-191 12485959-1 2003 7-Ethyl-10-hydroxycamptothecin (SN-38), an active metabolite of antitumor agent irinotecan (CPT-11), is conjugated and detoxified to SN-38-glucuronide by UDP-glucuronosyltransferase (UGT) 1A1. Irinotecan 80-90 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 154-191 12485959-1 2003 7-Ethyl-10-hydroxycamptothecin (SN-38), an active metabolite of antitumor agent irinotecan (CPT-11), is conjugated and detoxified to SN-38-glucuronide by UDP-glucuronosyltransferase (UGT) 1A1. Irinotecan 92-98 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 154-191 12485959-2 2003 Genetic polymorphisms in UGT1A1 are thought to contribute to severe diarrhea and/or leukopenia caused by CPT-11. Irinotecan 105-111 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 25-31 12485959-4 2003 However, little information is available on the influence of UGT1A1 polymorphisms in the coding region on the SN-38 glucuronidation activity. Irinotecan 110-115 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 61-67 12485959-7 2003 WT UGT1A1 catalyzed SN-38 glucuronidation with an apparent K(m) value of 11.5 microM, whereas those of G71R, P229Q, and Y486D were 14.0, 18.0, and 63.5 microM, respectively. Irinotecan 20-25 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 3-9 12485959-10 2003 The decreased SN-38 glucuronidation efficiency ratio of G71R and P229Q could be critical in combination with other polymorphisms in the UGT1A1 gene. Irinotecan 14-19 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 136-142 12533678-1 2003 Breast cancer resistance protein (BCRP), an ATP-binding cassette transporter, confers resistance to a series of anticancer agents such as SN-38, mitoxantrone, and topotecan. Irinotecan 138-143 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 0-32 12533678-1 2003 Breast cancer resistance protein (BCRP), an ATP-binding cassette transporter, confers resistance to a series of anticancer agents such as SN-38, mitoxantrone, and topotecan. Irinotecan 138-143 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 34-38 12533678-13 2003 Reversal of SN-38 and mitoxantrone resistance in K562/BCRP cells by TAG-139 was 5-fold stronger than that by estrone. Irinotecan 12-17 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 54-58 12607608-8 2002 The results suggest that liver and gastrointestinal tract with carboxylesterase-2 are likely the most important sites of conversion of irinotecan to the active metabolite SN-38, but carboxylesterase-2 within the other tissues may be contributive to this process. Irinotecan 135-145 carboxylesterase 2 Homo sapiens 63-81 12607608-8 2002 The results suggest that liver and gastrointestinal tract with carboxylesterase-2 are likely the most important sites of conversion of irinotecan to the active metabolite SN-38, but carboxylesterase-2 within the other tissues may be contributive to this process. Irinotecan 171-176 carboxylesterase 2 Homo sapiens 63-81 12384538-0 2002 Cyclooxygenase-2 inhibition with celecoxib enhances antitumor efficacy and reduces diarrhea side effect of CPT-11. Irinotecan 107-113 prostaglandin-endoperoxide synthase 2 Homo sapiens 0-16 12464801-4 2002 We also investigated the relationship between PBREM-(TA)n haplotypes and the glucuronidation rate of the UGT1A1 substrate SN-38. Irinotecan 122-127 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 105-111 12218061-3 2002 Here we demonstrate that the first 4-8 N-terminal amino acids of Smac/DIABLO fused to the Drosophila antennapaedia penetratin sequence, a carrier peptide, enhance the induction of apoptosis and long term antiproliferative effects of diverse antineoplastic agents including paclitaxel, etoposide, 7-ethyl-10-hydroxycamptothecin (SN-38), and doxorubicin in MCF-7 breast cancer cells. Irinotecan 296-326 diablo Drosophila melanogaster 65-69 12218061-3 2002 Here we demonstrate that the first 4-8 N-terminal amino acids of Smac/DIABLO fused to the Drosophila antennapaedia penetratin sequence, a carrier peptide, enhance the induction of apoptosis and long term antiproliferative effects of diverse antineoplastic agents including paclitaxel, etoposide, 7-ethyl-10-hydroxycamptothecin (SN-38), and doxorubicin in MCF-7 breast cancer cells. Irinotecan 296-326 diablo Drosophila melanogaster 70-76 12218061-3 2002 Here we demonstrate that the first 4-8 N-terminal amino acids of Smac/DIABLO fused to the Drosophila antennapaedia penetratin sequence, a carrier peptide, enhance the induction of apoptosis and long term antiproliferative effects of diverse antineoplastic agents including paclitaxel, etoposide, 7-ethyl-10-hydroxycamptothecin (SN-38), and doxorubicin in MCF-7 breast cancer cells. Irinotecan 328-333 diablo Drosophila melanogaster 65-69 12218061-3 2002 Here we demonstrate that the first 4-8 N-terminal amino acids of Smac/DIABLO fused to the Drosophila antennapaedia penetratin sequence, a carrier peptide, enhance the induction of apoptosis and long term antiproliferative effects of diverse antineoplastic agents including paclitaxel, etoposide, 7-ethyl-10-hydroxycamptothecin (SN-38), and doxorubicin in MCF-7 breast cancer cells. Irinotecan 328-333 diablo Drosophila melanogaster 70-76 12482336-0 2002 Combination of 5-fluorouracil and irinotecan on modulation of thymidylate synthase and topoisomerase I expression and cell cycle regulation in human colon cancer LoVo cells: clinical relevance. Irinotecan 34-44 thymidylate synthetase Homo sapiens 62-82 12465401-0 2002 [A case of AFP-producing gastric cancer with multiple liver metastases responding to CPT-11 and cisplatin combination chemotherapy]. Irinotecan 85-91 alpha fetoprotein Homo sapiens 11-14 12465401-1 2002 A case of AFP-producing gastric cancer successfully treated with CPT-11 and cisplatin combined therapy is reported together with a review of the literature. Irinotecan 65-71 alpha fetoprotein Homo sapiens 10-13 12465401-8 2002 This result suggests that combination chemotherapy consisting of CPT-11 and cisplatin may be effective and safe for patients with AFP-producing gastric cancer with multiple liver metastases. Irinotecan 65-71 alpha fetoprotein Homo sapiens 130-133 12484020-0 2002 [A case of postoperative liver metastasis of alpha-fetoprotein producing gastric cancer successfully treated with intrahepatic chemotherapy (adriamycin, cisplatin, irinotecan hydrochloride)]. Irinotecan 164-188 alpha fetoprotein Homo sapiens 45-62 12484020-5 2002 Twenty cycles of intrahepatic arterial infusion with adriamycin, cisplatin, and irinotecan hydrochloride resulted in a 93% decrease in the liver metastases along with normalization of the serum AFP level. Irinotecan 80-104 alpha fetoprotein Homo sapiens 194-197 12370755-0 2002 Phosphorylation of chk1 at serine-345 affected by topoisomerase I poison SN-38. Irinotecan 73-78 checkpoint kinase 1 Homo sapiens 19-23 12370755-6 2002 Increased chk1 phosphorylation at Ser345 induced by SN-38 was accompanied by the observed G2 phase arrest in the A253 cell line, while significant downregulation of chk1 and cdc25C phosphorylation, which resulted in the abrogation of G2/M checkpoint arrest, was noted in FaDu cells at this timepoint. Irinotecan 52-57 checkpoint kinase 1 Homo sapiens 10-14 12370755-7 2002 These results suggest that alterations of chk1 signaling are associated with the response to topoisomerase I poison SN-38. Irinotecan 116-121 checkpoint kinase 1 Homo sapiens 42-46 12370755-9 2002 A deficiency in G2 arrest was observed in FaDu cells, suggesting endogenous hMLH1 protein expression is associated with the abrogation of G2/M arrest, subsequently with the response to topoisomerase I poison SN-38. Irinotecan 208-213 mutL homolog 1 Homo sapiens 76-81 12440157-3 2002 To increase the clinical usefulness of theanine, we examined its effects on the antitumor activity of cisplatin and irinotecan (CPT-11), which a known to be transported by the efflux system related to MRP. Irinotecan 116-126 ATP-binding cassette, sub-family C (CFTR/MRP), member 1 Mus musculus 201-204 12440157-3 2002 To increase the clinical usefulness of theanine, we examined its effects on the antitumor activity of cisplatin and irinotecan (CPT-11), which a known to be transported by the efflux system related to MRP. Irinotecan 128-134 ATP-binding cassette, sub-family C (CFTR/MRP), member 1 Mus musculus 201-204 12384538-6 2002 We also assessed the involvement of COX-2 in the pathogenesis of CPT-11-induced late diarrhea using a rat model. Irinotecan 65-71 cytochrome c oxidase II, mitochondrial Rattus norvegicus 36-41 12384541-5 2002 Combining TRAIL with either 5-fluorouracil (5-FU) or CPT-11 (irinotecan hydrochloride) produced a greatly enhanced antitumor effect over that of either agent alone, with 50% of the animals achieving complete tumor regression with a combination of TRAIL and CPT-11. Irinotecan 53-59 TNF superfamily member 10 Homo sapiens 247-252 12384541-5 2002 Combining TRAIL with either 5-fluorouracil (5-FU) or CPT-11 (irinotecan hydrochloride) produced a greatly enhanced antitumor effect over that of either agent alone, with 50% of the animals achieving complete tumor regression with a combination of TRAIL and CPT-11. Irinotecan 61-85 TNF superfamily member 10 Homo sapiens 10-15 12384541-5 2002 Combining TRAIL with either 5-fluorouracil (5-FU) or CPT-11 (irinotecan hydrochloride) produced a greatly enhanced antitumor effect over that of either agent alone, with 50% of the animals achieving complete tumor regression with a combination of TRAIL and CPT-11. Irinotecan 61-85 TNF superfamily member 10 Homo sapiens 247-252 12384541-5 2002 Combining TRAIL with either 5-fluorouracil (5-FU) or CPT-11 (irinotecan hydrochloride) produced a greatly enhanced antitumor effect over that of either agent alone, with 50% of the animals achieving complete tumor regression with a combination of TRAIL and CPT-11. Irinotecan 257-263 TNF superfamily member 10 Homo sapiens 10-15 12384541-9 2002 Tumors treated with CPT-11 showed increased membrane expression of DR5, suggesting that CPT-11 may increase sensitivity to TRAIL by up-regulation of DR5. Irinotecan 20-26 TNF receptor superfamily member 10b Homo sapiens 67-70 12384541-9 2002 Tumors treated with CPT-11 showed increased membrane expression of DR5, suggesting that CPT-11 may increase sensitivity to TRAIL by up-regulation of DR5. Irinotecan 20-26 TNF superfamily member 10 Homo sapiens 123-128 12384541-9 2002 Tumors treated with CPT-11 showed increased membrane expression of DR5, suggesting that CPT-11 may increase sensitivity to TRAIL by up-regulation of DR5. Irinotecan 20-26 TNF receptor superfamily member 10b Homo sapiens 149-152 12384541-9 2002 Tumors treated with CPT-11 showed increased membrane expression of DR5, suggesting that CPT-11 may increase sensitivity to TRAIL by up-regulation of DR5. Irinotecan 88-94 TNF receptor superfamily member 10b Homo sapiens 67-70 12384541-9 2002 Tumors treated with CPT-11 showed increased membrane expression of DR5, suggesting that CPT-11 may increase sensitivity to TRAIL by up-regulation of DR5. Irinotecan 88-94 TNF superfamily member 10 Homo sapiens 123-128 12384541-9 2002 Tumors treated with CPT-11 showed increased membrane expression of DR5, suggesting that CPT-11 may increase sensitivity to TRAIL by up-regulation of DR5. Irinotecan 88-94 TNF receptor superfamily member 10b Homo sapiens 149-152 12369998-3 2002 When overexpressed in cell lines, ABCG2 has the ability to confer high levels of resistance to anthracyclines, mitoxantrone, bisantrene, and the camptothecins topotecan and SN-38. Irinotecan 173-178 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 34-39 12209584-0 2002 Cellular effects of CPT-11 on colon carcinoma cells: dependence on p53 and hMLH1 status. Irinotecan 20-26 tumor protein p53 Homo sapiens 67-70 12237777-3 2002 An approach to achieve tumour specific activation of CPT-11 is to transduce the cDNA encoding carboxylesterase into tumour cells. Irinotecan 53-59 carboxylesterase 2 Homo sapiens 94-110 12237777-11 2002 Importantly, in combination with CPT-11 both recombinant carboxylesterase proteins exerted strong antiproliferative effects on human colon cancer cells. Irinotecan 33-39 carboxylesterase 2 Homo sapiens 57-73 12237778-8 2002 In vivo experiments in well-established 2780DX8 human tumour xenografts demonstrated that the growth inhibition induced by CPT-11 was more affected by breast cancer resistance protein expression than that of DX-8951f. Irinotecan 123-129 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 151-183 12209584-0 2002 Cellular effects of CPT-11 on colon carcinoma cells: dependence on p53 and hMLH1 status. Irinotecan 20-26 mutL homolog 1 Homo sapiens 75-80 12181297-8 2002 PAF-induced (10 microM) Cl- secretion was inhibited by carboxy-PTIO (200 microM) and ODQ (10 microM), whereas irinotecan-induced (500 microM) Cl- secretion was not significantly inhibited by these drugs. Irinotecan 110-120 PCNA clamp associated factor Rattus norvegicus 0-3 12359059-6 2002 (2) An inhibitor of the nuclear transport of GSTpi, edible mushroom lectin (Agaricus bisporus lectin, ABL), increased the sensitivity of the cancer cells to DOX and CDDP, and partially to CPT-11. Irinotecan 188-194 ABL proto-oncogene 1, non-receptor tyrosine kinase Homo sapiens 102-105 12181437-0 2002 Common human UGT1A polymorphisms and the altered metabolism of irinotecan active metabolite 7-ethyl-10-hydroxycamptothecin (SN-38). Irinotecan 92-122 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 13-18 12181437-0 2002 Common human UGT1A polymorphisms and the altered metabolism of irinotecan active metabolite 7-ethyl-10-hydroxycamptothecin (SN-38). Irinotecan 124-129 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 13-18 12181437-3 2002 The purpose of this study was to determine whether common polymorphic UDP-glucuronosyltransferase (UGT) affects SN-38 glucuronidation. Irinotecan 112-117 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 70-97 12181437-3 2002 The purpose of this study was to determine whether common polymorphic UDP-glucuronosyltransferase (UGT) affects SN-38 glucuronidation. Irinotecan 112-117 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 99-102 12181437-5 2002 To assess the relative activity of UGT isoenzymes for SN-38, rates of formation of SN-38-G were monitored by liquid chromatography/mass spectrometry analysis and normalized by level of UGT cellular expression. Irinotecan 54-59 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 35-38 12181437-6 2002 Determination of intrinsic clearances predicts that hepatic UGT1A1 and UGT1A9 and the extrahepatic UGT1A7 are major components in SN-38-G formation, whereas a minor role is suggested for UGT1A6, UGT1A8, and UGT1A10. Irinotecan 130-135 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 60-66 12181437-6 2002 Determination of intrinsic clearances predicts that hepatic UGT1A1 and UGT1A9 and the extrahepatic UGT1A7 are major components in SN-38-G formation, whereas a minor role is suggested for UGT1A6, UGT1A8, and UGT1A10. Irinotecan 130-135 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 71-77 12181437-6 2002 Determination of intrinsic clearances predicts that hepatic UGT1A1 and UGT1A9 and the extrahepatic UGT1A7 are major components in SN-38-G formation, whereas a minor role is suggested for UGT1A6, UGT1A8, and UGT1A10. Irinotecan 130-135 UDP glucuronosyltransferase family 1 member A7 Homo sapiens 99-105 12181437-7 2002 In support of the involvement of UGT1A9, a strong coefficient of correlation was observed in the glucuronidation of SN-38 and a substrate, mainly glucuronidate, by UGT1A9 (flavopiridol) by human liver microsomes (coefficient of correlation, 0.905; p = 0.002). Irinotecan 116-121 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 33-39 12181437-7 2002 In support of the involvement of UGT1A9, a strong coefficient of correlation was observed in the glucuronidation of SN-38 and a substrate, mainly glucuronidate, by UGT1A9 (flavopiridol) by human liver microsomes (coefficient of correlation, 0.905; p = 0.002). Irinotecan 116-121 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 164-170 12181437-11 2002 Taken together, these data provide the evidence that molecular determinants of irinotecan response may include the UGT1A polymorphisms studied herein and common genetic variants of the hepatic UGT1A9 isoenzyme yet to be described. Irinotecan 79-89 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 115-120 12181437-11 2002 Taken together, these data provide the evidence that molecular determinants of irinotecan response may include the UGT1A polymorphisms studied herein and common genetic variants of the hepatic UGT1A9 isoenzyme yet to be described. Irinotecan 79-89 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 193-199 12380945-4 2002 Irinotecan (CPT-II, Camptosar) has demonstrated efficacy in the treatment of small-cell lung cancer (SCLC). Irinotecan 0-10 carnitine palmitoyltransferase 2 Homo sapiens 12-18 12189228-2 2002 In this study, we determined the effect of SJW on the metabolism of irinotecan, a pro-drug of SN-38 and a known substrate for CYP3A4. Irinotecan 68-78 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 126-132 12171891-5 2002 This scenario raises the possibility that local conversion of irinotecan to SN-38 by CES2 in tumor tissues might occur. Irinotecan 62-72 carboxylesterase 2 Homo sapiens 85-89 12171891-0 2002 Human carboxylesterase 2 is commonly expressed in tumor tissue and is correlated with activation of irinotecan. Irinotecan 100-110 carboxylesterase 2 Homo sapiens 6-24 12171891-5 2002 This scenario raises the possibility that local conversion of irinotecan to SN-38 by CES2 in tumor tissues might occur. Irinotecan 76-81 carboxylesterase 2 Homo sapiens 85-89 12171891-10 2002 Liver microsomal CES2 protein expression was significantly correlated with irinotecan activation to SN-38 (R(s) = 0.70; P = 0.007). Irinotecan 75-85 carboxylesterase 2 Homo sapiens 17-21 12171891-10 2002 Liver microsomal CES2 protein expression was significantly correlated with irinotecan activation to SN-38 (R(s) = 0.70; P = 0.007). Irinotecan 100-105 carboxylesterase 2 Homo sapiens 17-21 12171891-11 2002 This study confirms that CES2 is a key enzyme for irinotecan activation. Irinotecan 50-60 carboxylesterase 2 Homo sapiens 25-29 12171891-12 2002 Tumor CES2 expression may contribute to variable response to irinotecan chemotherapy for solid tumors. Irinotecan 61-71 carboxylesterase 2 Homo sapiens 6-10 12171903-0 2002 Irinotecan activation by human carboxylesterases in colorectal adenocarcinoma cells. Irinotecan 0-10 carboxylesterase 1 Homo sapiens 31-48 12171903-1 2002 Carboxylesterases play a critical role in the bioactivation of the anticancer prodrug irinotecan [7-ethyl-10-[4-(1-piperidino)-1-piperidino]carbonyloxy-camptothecin; CPT-11] into its active metabolite SN-38 (ethyl-10-hydroxy-camptothecin). Irinotecan 86-96 carboxylesterase 1 Homo sapiens 0-17 12171903-1 2002 Carboxylesterases play a critical role in the bioactivation of the anticancer prodrug irinotecan [7-ethyl-10-[4-(1-piperidino)-1-piperidino]carbonyloxy-camptothecin; CPT-11] into its active metabolite SN-38 (ethyl-10-hydroxy-camptothecin). Irinotecan 98-164 carboxylesterase 1 Homo sapiens 0-17 12171903-1 2002 Carboxylesterases play a critical role in the bioactivation of the anticancer prodrug irinotecan [7-ethyl-10-[4-(1-piperidino)-1-piperidino]carbonyloxy-camptothecin; CPT-11] into its active metabolite SN-38 (ethyl-10-hydroxy-camptothecin). Irinotecan 166-172 carboxylesterase 1 Homo sapiens 0-17 12171903-1 2002 Carboxylesterases play a critical role in the bioactivation of the anticancer prodrug irinotecan [7-ethyl-10-[4-(1-piperidino)-1-piperidino]carbonyloxy-camptothecin; CPT-11] into its active metabolite SN-38 (ethyl-10-hydroxy-camptothecin). Irinotecan 201-206 carboxylesterase 1 Homo sapiens 0-17 12171903-2 2002 We reported recently that human carboxylesterase-2 (hCE-2) is a higher-affinity, higher-velocity enzyme for irinotecan hydrolysis when compared with hCE-1. Irinotecan 108-118 carboxylesterase 2 Homo sapiens 32-50 12171903-2 2002 We reported recently that human carboxylesterase-2 (hCE-2) is a higher-affinity, higher-velocity enzyme for irinotecan hydrolysis when compared with hCE-1. Irinotecan 108-118 carboxylesterase 2 Homo sapiens 52-57 12171903-4 2002 Extracts of HT29 cells transfected with hCE-2 exhibited significantly higher irinotecan hydrolysis (5.2 pmol/mg protein/hr) than hCE-1 (1.0 pmol/mg protein/hr). Irinotecan 77-87 carboxylesterase 2 Homo sapiens 40-45 12171903-5 2002 HT29 cells over-expressing hCE-2 were more sensitive to the toxic effects of irinotecan than cells expressing hCE-1 (EC50 = 0.3 micro M and 6.8 micro M, respectively). Irinotecan 77-87 carboxylesterase 2 Homo sapiens 27-32 12171903-6 2002 Our data further support the notion that hCE-2 plays a substantial role in irinotecan activation in human tissue at relevant pharmacologic concentrations. Irinotecan 75-85 carboxylesterase 2 Homo sapiens 41-46 12199631-2 2002 The hepatic metabolism and biliary secretion of irinotecan (CPT-11, Camptosar) and metabolites is complex and involves cytochrome P450 isoenzymes, carboxylesterases, glucuronosyltransferase, and the ATP-dependent export pumps MRP-2 and MXR. Irinotecan 48-58 ATP binding cassette subfamily C member 2 Homo sapiens 226-231 12199631-2 2002 The hepatic metabolism and biliary secretion of irinotecan (CPT-11, Camptosar) and metabolites is complex and involves cytochrome P450 isoenzymes, carboxylesterases, glucuronosyltransferase, and the ATP-dependent export pumps MRP-2 and MXR. Irinotecan 48-58 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 236-239 12199631-2 2002 The hepatic metabolism and biliary secretion of irinotecan (CPT-11, Camptosar) and metabolites is complex and involves cytochrome P450 isoenzymes, carboxylesterases, glucuronosyltransferase, and the ATP-dependent export pumps MRP-2 and MXR. Irinotecan 60-66 ATP binding cassette subfamily C member 2 Homo sapiens 226-231 12199631-2 2002 The hepatic metabolism and biliary secretion of irinotecan (CPT-11, Camptosar) and metabolites is complex and involves cytochrome P450 isoenzymes, carboxylesterases, glucuronosyltransferase, and the ATP-dependent export pumps MRP-2 and MXR. Irinotecan 60-66 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 236-239 12199631-2 2002 The hepatic metabolism and biliary secretion of irinotecan (CPT-11, Camptosar) and metabolites is complex and involves cytochrome P450 isoenzymes, carboxylesterases, glucuronosyltransferase, and the ATP-dependent export pumps MRP-2 and MXR. Irinotecan 68-77 ATP binding cassette subfamily C member 2 Homo sapiens 226-231 12199631-2 2002 The hepatic metabolism and biliary secretion of irinotecan (CPT-11, Camptosar) and metabolites is complex and involves cytochrome P450 isoenzymes, carboxylesterases, glucuronosyltransferase, and the ATP-dependent export pumps MRP-2 and MXR. Irinotecan 68-77 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 236-239 12479221-1 2002 Breast cancer resistance protein (BCRP) confers multidrug resistance to cancer cells against agents such as SN-38 (an active metabolite of irinotecan), mitoxantrone, and topotecan. Irinotecan 108-113 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 0-32 12124325-3 2002 One of the genes markedly up-regulated during cell cycle arrest by SN-38 and suppressed during apoptosis by SN-38 followed by flavopiridol in Hct116 cells is Drg1. Irinotecan 67-72 developmentally regulated GTP binding protein 1 Homo sapiens 158-162 12124325-3 2002 One of the genes markedly up-regulated during cell cycle arrest by SN-38 and suppressed during apoptosis by SN-38 followed by flavopiridol in Hct116 cells is Drg1. Irinotecan 108-113 developmentally regulated GTP binding protein 1 Homo sapiens 158-162 12124325-4 2002 We found that Drg1 had profound effects on SN-38 sensitivity. Irinotecan 43-48 developmentally regulated GTP binding protein 1 Homo sapiens 14-18 12124325-5 2002 Inhibition of endogenous Drg1 expression in Hct116 cells by stable expression of an antisense (AS) Drg1 cDNA increased the sensitivity of cells to undergo apoptosis by SN-38. Irinotecan 168-173 developmentally regulated GTP binding protein 1 Homo sapiens 25-29 12124325-5 2002 Inhibition of endogenous Drg1 expression in Hct116 cells by stable expression of an antisense (AS) Drg1 cDNA increased the sensitivity of cells to undergo apoptosis by SN-38. Irinotecan 168-173 developmentally regulated GTP binding protein 1 Homo sapiens 99-103 12124325-6 2002 Clonogenic and apoptosis assays with AS Drg1-expressing cells showed a 2-fold decrease in the IC50 and a 4-5-fold increase in induction of apoptosis with SN-38. Irinotecan 154-159 developmentally regulated GTP binding protein 1 Homo sapiens 40-44 12124325-7 2002 Conversely, the forced expression of Drg1 in SW620 cells increased the resistance of these cells to SN-38-induced apoptosis by 2-5-fold. Irinotecan 100-105 developmentally regulated GTP binding protein 1 Homo sapiens 37-41 12124325-8 2002 Moreover, when xenografted in mice, AS Drg1-expressing Hct116 cells were 3-fold more sensitive to CPT-11 as compared with vector transfected Hct116 cells. Irinotecan 98-104 developmentally regulated GTP binding protein 1 Mus musculus 39-43 12124325-9 2002 Similarly, tumors established from Drg1 overexpressing SW620 cells were more resistant to CPT-11 as compared with tumors established from vector-transfected SW620 cells in mice. Irinotecan 90-96 developmentally regulated GTP binding protein 1 Homo sapiens 35-39 12124325-10 2002 Taken together, our data suggest that Drg1 is a novel gene that plays a direct role in resistance to CPT-11. Irinotecan 101-107 developmentally regulated GTP binding protein 1 Homo sapiens 38-42 12124325-11 2002 Inhibition of Drg1 may provide a new means to increase the sensitivity of colon cancer cells to CPT-11. Irinotecan 96-102 developmentally regulated GTP binding protein 1 Homo sapiens 14-18 12065434-0 2002 Intestinal transport of irinotecan in Caco-2 cells and MDCK II cells overexpressing efflux transporters Pgp, cMOAT, and MRP1. Irinotecan 24-34 PGP Canis lupus familiaris 104-107 12065434-0 2002 Intestinal transport of irinotecan in Caco-2 cells and MDCK II cells overexpressing efflux transporters Pgp, cMOAT, and MRP1. Irinotecan 24-34 ATP binding cassette subfamily C member 1 Canis lupus familiaris 120-124 12143255-5 2002 After 2-4 months of CPT-11-based chemotherapy, significant increase in CD3+CD4+ cell numbers was observed in 8 patients who had initial CD3+CD4+ counts of less than 600 per microL (358 +/- 154 vs. 652 +/- 319 cells per microL, Wilcoxon test, P < 0.01), while in patients with higher initial CD3+CD4+ counts a trend for decrease was observed during therapy. Irinotecan 20-26 CD4 molecule Homo sapiens 75-78 12143255-5 2002 After 2-4 months of CPT-11-based chemotherapy, significant increase in CD3+CD4+ cell numbers was observed in 8 patients who had initial CD3+CD4+ counts of less than 600 per microL (358 +/- 154 vs. 652 +/- 319 cells per microL, Wilcoxon test, P < 0.01), while in patients with higher initial CD3+CD4+ counts a trend for decrease was observed during therapy. Irinotecan 20-26 CD4 molecule Homo sapiens 140-143 12143255-5 2002 After 2-4 months of CPT-11-based chemotherapy, significant increase in CD3+CD4+ cell numbers was observed in 8 patients who had initial CD3+CD4+ counts of less than 600 per microL (358 +/- 154 vs. 652 +/- 319 cells per microL, Wilcoxon test, P < 0.01), while in patients with higher initial CD3+CD4+ counts a trend for decrease was observed during therapy. Irinotecan 20-26 CD4 molecule Homo sapiens 140-143 12143255-8 2002 CPT-11-based therapy seems to ameliorate CD3+CD4+ lymphocytopenia, possibly by neutralizing the immunosuppressive effects of uncontrolled tumor growth. Irinotecan 0-6 CD4 molecule Homo sapiens 45-48 12118026-8 2002 CONCLUSION: Inhibition of CYP3A4 in cancer patients treated with irinotecan leads to significantly increased formation of SN-38. Irinotecan 65-75 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 26-32 12118026-8 2002 CONCLUSION: Inhibition of CYP3A4 in cancer patients treated with irinotecan leads to significantly increased formation of SN-38. Irinotecan 122-127 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 26-32 12065434-9 2002 Furthermore, the secretory efflux P(eff) of CPT-11 was significantly decreased by Pgp inhibitors, elacridar (GF120918) (IC50 = 0.38 +/- 0.06 microM) and verapamil (IC(50) = 234 +/- 48 microM) in MDCK II/Pgp cells and by cMOAT inhibitor 3-([(3-(2-[7-chloro-2-quinolinyl]ethyl)phenyl]-[(3-dimethylamino-3-oxoprphyl)-thio)-methyl]-thio) propanoic acid (MK571) (IC50) = 469 +/- 60 micro;M) in MDCK II/cMOAT cells. Irinotecan 44-50 PGP Canis lupus familiaris 82-85 12065434-9 2002 Furthermore, the secretory efflux P(eff) of CPT-11 was significantly decreased by Pgp inhibitors, elacridar (GF120918) (IC50 = 0.38 +/- 0.06 microM) and verapamil (IC(50) = 234 +/- 48 microM) in MDCK II/Pgp cells and by cMOAT inhibitor 3-([(3-(2-[7-chloro-2-quinolinyl]ethyl)phenyl]-[(3-dimethylamino-3-oxoprphyl)-thio)-methyl]-thio) propanoic acid (MK571) (IC50) = 469 +/- 60 micro;M) in MDCK II/cMOAT cells. Irinotecan 44-50 PGP Canis lupus familiaris 203-206 12065434-10 2002 Overall, the current study suggests that Pgp and cMOAT are capable of mediating the efflux of CPT-11 in vitro. Irinotecan 94-100 PGP Canis lupus familiaris 41-44 12063551-2 2002 The combination treatment of GEM 231 and irinotecan produced enhanced and prolonged tumor-growth inhibition, compared with irinotecan monotherapy, against human colon (HCT-116), pancreas (Panc-1), prostate (PC3) and lung (SKMES) tumors in mice. Irinotecan 41-51 proprotein convertase subtilisin/kexin type 1 Homo sapiens 207-210 12094251-8 2002 After a 7-day exposure to anti-CD32, the recovery of ALL cells cocultured with stroma was reduced to 5.5% +/- 2.8% of control values in 380 cells (n = 14), 19.4% +/- 6.1% (n = 8) in RS4;11, and 4.0% +/- 1.3% (n = 6) in KOPN55bi. Irinotecan 182-185 Fc gamma receptor IIa Homo sapiens 31-35 12019202-9 2002 In conclusion, potential clinical interactions with the metabolism of irinotecan are likely to be important for vinorelbine, which strongly inhibits irinotecan catabolism by CYP3A4 at clinically relevant concentrations, but not for the other drugs, which exert an effect at concentrations not achievable in patients. Irinotecan 70-80 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 174-180 12019202-9 2002 In conclusion, potential clinical interactions with the metabolism of irinotecan are likely to be important for vinorelbine, which strongly inhibits irinotecan catabolism by CYP3A4 at clinically relevant concentrations, but not for the other drugs, which exert an effect at concentrations not achievable in patients. Irinotecan 149-159 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 174-180 12479221-1 2002 Breast cancer resistance protein (BCRP) confers multidrug resistance to cancer cells against agents such as SN-38 (an active metabolite of irinotecan), mitoxantrone, and topotecan. Irinotecan 108-113 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 34-38 12479221-1 2002 Breast cancer resistance protein (BCRP) confers multidrug resistance to cancer cells against agents such as SN-38 (an active metabolite of irinotecan), mitoxantrone, and topotecan. Irinotecan 139-149 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 0-32 12479221-1 2002 Breast cancer resistance protein (BCRP) confers multidrug resistance to cancer cells against agents such as SN-38 (an active metabolite of irinotecan), mitoxantrone, and topotecan. Irinotecan 139-149 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 34-38 12479221-11 2002 Therefore, people with C376T and/or C421A polymorphisms may express low amounts of BCRP, and this low BCRP expression might result in hypersensitivity of normal cells to such anticancer drugs as irinotecan and mitoxantrone. Irinotecan 195-205 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 83-87 12479221-11 2002 Therefore, people with C376T and/or C421A polymorphisms may express low amounts of BCRP, and this low BCRP expression might result in hypersensitivity of normal cells to such anticancer drugs as irinotecan and mitoxantrone. Irinotecan 195-205 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 102-106 11888671-0 2002 Loss of one allele of the p53 gene in the lens epithelial tumor in transgenic mice suppresses apoptosis induced by a topoisomerase I inhibitor (CPT-11). Irinotecan 144-150 transformation related protein 53, pseudogene Mus musculus 26-29 11888671-6 2002 In addition, it was found that CPT-11 could also induce apoptosis via a p53-independent pathway. Irinotecan 31-37 transformation related protein 53, pseudogene Mus musculus 72-75 12032863-0 2002 Enhanced tumor killing by Apo2L/TRAIL and CPT-11 co-treatment is associated with p21 cleavage and differential regulation of Apo2L/TRAIL ligand and its receptors. Irinotecan 42-48 H3 histone pseudogene 16 Homo sapiens 81-84 12032863-0 2002 Enhanced tumor killing by Apo2L/TRAIL and CPT-11 co-treatment is associated with p21 cleavage and differential regulation of Apo2L/TRAIL ligand and its receptors. Irinotecan 42-48 TNF superfamily member 10 Homo sapiens 125-130 12032863-0 2002 Enhanced tumor killing by Apo2L/TRAIL and CPT-11 co-treatment is associated with p21 cleavage and differential regulation of Apo2L/TRAIL ligand and its receptors. Irinotecan 42-48 TNF superfamily member 10 Homo sapiens 131-136 12032863-1 2002 Apo2L/TRAIL exhibits enhanced apoptotic activity in tumor xenograft models when used in combination with the topoisomerase 1 inhibitor CPT-11. Irinotecan 135-141 TNF superfamily member 10 Homo sapiens 0-5 12032863-1 2002 Apo2L/TRAIL exhibits enhanced apoptotic activity in tumor xenograft models when used in combination with the topoisomerase 1 inhibitor CPT-11. Irinotecan 135-141 TNF superfamily member 10 Homo sapiens 6-11 12032863-4 2002 Upregulation of decoy receptors by CPT-11 was partially inhibited by co-administration of Apo2L/TRAIL. Irinotecan 35-41 TNF superfamily member 10 Homo sapiens 90-95 12032863-4 2002 Upregulation of decoy receptors by CPT-11 was partially inhibited by co-administration of Apo2L/TRAIL. Irinotecan 35-41 TNF superfamily member 10 Homo sapiens 96-101 12032863-5 2002 CPT-11 treatment resulted in accumulation of cells at G(2)M-phase and correlated with a substantial increase in the protein levels of the cyclin-dependent kinase inhibitor p21. Irinotecan 0-6 H3 histone pseudogene 16 Homo sapiens 172-175 12032863-6 2002 However, cells co-treated with CPT-11 and Apo2L/TRAIL, or pretreated with CPT-11 for up to 24 h followed by 2 h Apo2L/TRAIL, resulted in a caspase-dependent degradation of p21, reversal of G(2)-M phase arrest with a concomitant increase in apoptosis. Irinotecan 31-37 TNF superfamily member 10 Homo sapiens 112-117 12032863-6 2002 However, cells co-treated with CPT-11 and Apo2L/TRAIL, or pretreated with CPT-11 for up to 24 h followed by 2 h Apo2L/TRAIL, resulted in a caspase-dependent degradation of p21, reversal of G(2)-M phase arrest with a concomitant increase in apoptosis. Irinotecan 31-37 TNF superfamily member 10 Homo sapiens 118-123 12032863-6 2002 However, cells co-treated with CPT-11 and Apo2L/TRAIL, or pretreated with CPT-11 for up to 24 h followed by 2 h Apo2L/TRAIL, resulted in a caspase-dependent degradation of p21, reversal of G(2)-M phase arrest with a concomitant increase in apoptosis. Irinotecan 31-37 H3 histone pseudogene 16 Homo sapiens 172-175 12032863-6 2002 However, cells co-treated with CPT-11 and Apo2L/TRAIL, or pretreated with CPT-11 for up to 24 h followed by 2 h Apo2L/TRAIL, resulted in a caspase-dependent degradation of p21, reversal of G(2)-M phase arrest with a concomitant increase in apoptosis. Irinotecan 74-80 TNF superfamily member 10 Homo sapiens 118-123 12032863-6 2002 However, cells co-treated with CPT-11 and Apo2L/TRAIL, or pretreated with CPT-11 for up to 24 h followed by 2 h Apo2L/TRAIL, resulted in a caspase-dependent degradation of p21, reversal of G(2)-M phase arrest with a concomitant increase in apoptosis. Irinotecan 74-80 H3 histone pseudogene 16 Homo sapiens 172-175 12032863-8 2002 These data indicate that the up-regulation of Apo2L/TRAIL ligand and its death receptors as well as cleavage of p21 protein in the Apo2L/TRAIL plus CPT-11 treatment contributes to the positive cooperation between these agents in enhancing tumor cell apoptosis. Irinotecan 148-154 TNF superfamily member 10 Homo sapiens 46-51 12032863-8 2002 These data indicate that the up-regulation of Apo2L/TRAIL ligand and its death receptors as well as cleavage of p21 protein in the Apo2L/TRAIL plus CPT-11 treatment contributes to the positive cooperation between these agents in enhancing tumor cell apoptosis. Irinotecan 148-154 TNF superfamily member 10 Homo sapiens 52-57 12032863-8 2002 These data indicate that the up-regulation of Apo2L/TRAIL ligand and its death receptors as well as cleavage of p21 protein in the Apo2L/TRAIL plus CPT-11 treatment contributes to the positive cooperation between these agents in enhancing tumor cell apoptosis. Irinotecan 148-154 H3 histone pseudogene 16 Homo sapiens 112-115 12032863-8 2002 These data indicate that the up-regulation of Apo2L/TRAIL ligand and its death receptors as well as cleavage of p21 protein in the Apo2L/TRAIL plus CPT-11 treatment contributes to the positive cooperation between these agents in enhancing tumor cell apoptosis. Irinotecan 148-154 TNF superfamily member 10 Homo sapiens 131-136 12032863-8 2002 These data indicate that the up-regulation of Apo2L/TRAIL ligand and its death receptors as well as cleavage of p21 protein in the Apo2L/TRAIL plus CPT-11 treatment contributes to the positive cooperation between these agents in enhancing tumor cell apoptosis. Irinotecan 148-154 TNF superfamily member 10 Homo sapiens 137-142 12006545-0 2002 Overexpression of PTEN increases sensitivity to SN-38, an active metabolite of the topoisomerase I inhibitor irinotecan, in ovarian cancer cells. Irinotecan 48-53 phosphatase and tensin homolog Homo sapiens 18-22 12006545-0 2002 Overexpression of PTEN increases sensitivity to SN-38, an active metabolite of the topoisomerase I inhibitor irinotecan, in ovarian cancer cells. Irinotecan 109-119 phosphatase and tensin homolog Homo sapiens 18-22 12006545-7 2002 In vitro examination of sensitivity to anticancer agents showed that the 50% growth-inhibitory concentration value for irinotecan metabolite (SN-38) in SHIN-3/PTEN was 800 nM, a 6.6-fold higher sensitivity compared with that of the control (5300 nM). Irinotecan 119-129 phosphatase and tensin homolog Homo sapiens 159-163 12006545-7 2002 In vitro examination of sensitivity to anticancer agents showed that the 50% growth-inhibitory concentration value for irinotecan metabolite (SN-38) in SHIN-3/PTEN was 800 nM, a 6.6-fold higher sensitivity compared with that of the control (5300 nM). Irinotecan 142-147 phosphatase and tensin homolog Homo sapiens 159-163 12006545-9 2002 The percentage of apoptotic cells in SHIN-3/PTEN was 16.6 +/- 0.7% 24 h after addition of SN-38, a significant increase over controls (8.6 +/- 0.9%; P < 0.01). Irinotecan 90-95 phosphatase and tensin homolog Homo sapiens 44-48 12006545-11 2002 CONCLUSIONS: These results indicate that high PTEN expression enhances the sensitivity of ovarian cancer cells to irinotecan and the induction of apoptosis and the suppression of topoisomerase I activity in cancer cells are suggested as possible mechanisms attributable to high PTEN expression. Irinotecan 114-124 phosphatase and tensin homolog Homo sapiens 46-50 11956509-0 2002 Cholinergic toxic syndrome by the anticancer drug irinotecan: acetylcholinesterase does not play a major role. Irinotecan 50-60 acetylcholinesterase (Cartwright blood group) Homo sapiens 62-82 12036456-2 2002 It is converted by carboxyesterase to an active metabolite, SN-38, which is further conjugated and detoxified to SN-38-glucuronide by UDP-glucuronosyltransferase (UGT). Irinotecan 60-65 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 134-161 12036456-2 2002 It is converted by carboxyesterase to an active metabolite, SN-38, which is further conjugated and detoxified to SN-38-glucuronide by UDP-glucuronosyltransferase (UGT). Irinotecan 60-65 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 163-166 12036456-4 2002 All the patients had received irinotecan-containing chemotherapy and were evaluated to see whether the variant UGT1A7 genotype would increase the likelihood of severe toxicity of irinotecan consisting of grade 4 leukopenia and/or grade 3 or more diarrhea. Irinotecan 179-189 UDP glucuronosyltransferase family 1 member A7 Homo sapiens 111-117 11914914-2 2002 We studied the role of P-glycoprotein (P-gp) in in vivo biliary excretion of CPT-11, SN-38 and SN-38G in mice lacking mdr1-type P-gp [ mdr1a/1b(-/-)] in the presence of the multidrug resistance (MDR) reversal agent, PSC833. Irinotecan 77-83 phosphoglycolate phosphatase Mus musculus 39-43 11914914-2 2002 We studied the role of P-glycoprotein (P-gp) in in vivo biliary excretion of CPT-11, SN-38 and SN-38G in mice lacking mdr1-type P-gp [ mdr1a/1b(-/-)] in the presence of the multidrug resistance (MDR) reversal agent, PSC833. Irinotecan 95-100 phosphoglycolate phosphatase Mus musculus 39-43 11914914-6 2002 This also implied a major role of other undetermined non-P-gp-mediated mechanism(s) for hepatic transport of CPT-11, which was inhibited by PSC833 (1.8+/-0.8% with PSC833, 6.6+/-0.6% without PSC833) in mdr1a/1b(-/-) mice. Irinotecan 109-115 phosphoglycolate phosphatase Mus musculus 57-61 11914914-6 2002 This also implied a major role of other undetermined non-P-gp-mediated mechanism(s) for hepatic transport of CPT-11, which was inhibited by PSC833 (1.8+/-0.8% with PSC833, 6.6+/-0.6% without PSC833) in mdr1a/1b(-/-) mice. Irinotecan 109-115 ATP-binding cassette, sub-family B (MDR/TAP), member 1A Mus musculus 202-207 11967565-2 2002 We present the 2.5 A crystal structure of rabbit liver carboxylesterase (rCE), the most efficient enzyme known to activate CPT-11 in this manner, in complex with the leaving group 4PP. Irinotecan 123-129 liver carboxylesterase 1 Oryctolagus cuniculus 49-71 11914914-0 2002 Biliary transport of irinotecan and metabolites in normal and P-glycoprotein-deficient mice. Irinotecan 21-31 phosphoglycolate phosphatase Mus musculus 62-76 11956509-2 2002 This study investigated (1) the pattern of acetylcholinesterase activity in patients receiving treatment with irinotecan and (2) the relationship between acetylcholinesterase activity and plasma concentrations of irinotecan, 7-ethyl-10-hydroxycamptothecin (SN-38), and SN-38 glucuronide (SN-38G). Irinotecan 213-223 acetylcholinesterase (Cartwright blood group) Homo sapiens 154-174 11956509-9 2002 In vitro, the activity of acetylcholinesterase was inhibited by 100-micromol/L irinotecan (-24.8%) and markedly reduced by 1-micromol/L physostigmine (-86.7%), whereas neither SN-38 nor camptothecin had an effect. Irinotecan 79-89 acetylcholinesterase (Cartwright blood group) Homo sapiens 26-46 11956509-9 2002 In vitro, the activity of acetylcholinesterase was inhibited by 100-micromol/L irinotecan (-24.8%) and markedly reduced by 1-micromol/L physostigmine (-86.7%), whereas neither SN-38 nor camptothecin had an effect. Irinotecan 176-181 acetylcholinesterase (Cartwright blood group) Homo sapiens 26-46 11956509-1 2002 BACKGROUND: The anticancer drug irinotecan induces cholinergic side effects that are currently ascribed to the blockade of acetylcholinesterase. Irinotecan 32-42 acetylcholinesterase (Cartwright blood group) Homo sapiens 123-143 11956509-2 2002 This study investigated (1) the pattern of acetylcholinesterase activity in patients receiving treatment with irinotecan and (2) the relationship between acetylcholinesterase activity and plasma concentrations of irinotecan, 7-ethyl-10-hydroxycamptothecin (SN-38), and SN-38 glucuronide (SN-38G). Irinotecan 110-120 acetylcholinesterase (Cartwright blood group) Homo sapiens 43-63 11990699-2 2002 Recent studies have shown that irinotecan also undergoes oxidative metabolism by the P450 isozyme CYP3A4, leading to the formation of a minor inactive metabolite, 7-ethyl-10-[4-N-[(5-aminopentanoic acid)-1-piperidino]-carbonyloxy-camptothecin (APC). Irinotecan 31-41 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 98-104 11901092-6 2002 Furthermore, CPT-11 and SN-38 were suggested to be mechanism-based inactivators of CYP3A4. Irinotecan 13-19 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 83-89 11901092-6 2002 Furthermore, CPT-11 and SN-38 were suggested to be mechanism-based inactivators of CYP3A4. Irinotecan 24-29 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 83-89 11901092-9 2002 These results mean that CPT-11 and SN-38 interact with human P450 isoforms, such as CYP2A6, CYP2C9, and CYP3A4, in vitro and imply that the significant drug interactions involving CPT-11 may be caused by a mechanism-based inactivation of CYP3A4 by SN-38 as an active metabolite of CPT-11 rather than competitive inhibition. Irinotecan 24-30 cytochrome P450 family 2 subfamily A member 6 Homo sapiens 84-90 11901092-9 2002 These results mean that CPT-11 and SN-38 interact with human P450 isoforms, such as CYP2A6, CYP2C9, and CYP3A4, in vitro and imply that the significant drug interactions involving CPT-11 may be caused by a mechanism-based inactivation of CYP3A4 by SN-38 as an active metabolite of CPT-11 rather than competitive inhibition. Irinotecan 24-30 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 92-98 11901092-9 2002 These results mean that CPT-11 and SN-38 interact with human P450 isoforms, such as CYP2A6, CYP2C9, and CYP3A4, in vitro and imply that the significant drug interactions involving CPT-11 may be caused by a mechanism-based inactivation of CYP3A4 by SN-38 as an active metabolite of CPT-11 rather than competitive inhibition. Irinotecan 24-30 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 104-110 11901092-9 2002 These results mean that CPT-11 and SN-38 interact with human P450 isoforms, such as CYP2A6, CYP2C9, and CYP3A4, in vitro and imply that the significant drug interactions involving CPT-11 may be caused by a mechanism-based inactivation of CYP3A4 by SN-38 as an active metabolite of CPT-11 rather than competitive inhibition. Irinotecan 24-30 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 238-244 11901092-9 2002 These results mean that CPT-11 and SN-38 interact with human P450 isoforms, such as CYP2A6, CYP2C9, and CYP3A4, in vitro and imply that the significant drug interactions involving CPT-11 may be caused by a mechanism-based inactivation of CYP3A4 by SN-38 as an active metabolite of CPT-11 rather than competitive inhibition. Irinotecan 35-40 cytochrome P450 family 2 subfamily A member 6 Homo sapiens 84-90 11901092-9 2002 These results mean that CPT-11 and SN-38 interact with human P450 isoforms, such as CYP2A6, CYP2C9, and CYP3A4, in vitro and imply that the significant drug interactions involving CPT-11 may be caused by a mechanism-based inactivation of CYP3A4 by SN-38 as an active metabolite of CPT-11 rather than competitive inhibition. Irinotecan 35-40 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 92-98 11901092-9 2002 These results mean that CPT-11 and SN-38 interact with human P450 isoforms, such as CYP2A6, CYP2C9, and CYP3A4, in vitro and imply that the significant drug interactions involving CPT-11 may be caused by a mechanism-based inactivation of CYP3A4 by SN-38 as an active metabolite of CPT-11 rather than competitive inhibition. Irinotecan 35-40 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 104-110 11901092-9 2002 These results mean that CPT-11 and SN-38 interact with human P450 isoforms, such as CYP2A6, CYP2C9, and CYP3A4, in vitro and imply that the significant drug interactions involving CPT-11 may be caused by a mechanism-based inactivation of CYP3A4 by SN-38 as an active metabolite of CPT-11 rather than competitive inhibition. Irinotecan 35-40 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 238-244 11901092-9 2002 These results mean that CPT-11 and SN-38 interact with human P450 isoforms, such as CYP2A6, CYP2C9, and CYP3A4, in vitro and imply that the significant drug interactions involving CPT-11 may be caused by a mechanism-based inactivation of CYP3A4 by SN-38 as an active metabolite of CPT-11 rather than competitive inhibition. Irinotecan 180-186 cytochrome P450 family 2 subfamily A member 6 Homo sapiens 84-90 11901092-9 2002 These results mean that CPT-11 and SN-38 interact with human P450 isoforms, such as CYP2A6, CYP2C9, and CYP3A4, in vitro and imply that the significant drug interactions involving CPT-11 may be caused by a mechanism-based inactivation of CYP3A4 by SN-38 as an active metabolite of CPT-11 rather than competitive inhibition. Irinotecan 180-186 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 92-98 11901092-9 2002 These results mean that CPT-11 and SN-38 interact with human P450 isoforms, such as CYP2A6, CYP2C9, and CYP3A4, in vitro and imply that the significant drug interactions involving CPT-11 may be caused by a mechanism-based inactivation of CYP3A4 by SN-38 as an active metabolite of CPT-11 rather than competitive inhibition. Irinotecan 180-186 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 104-110 11901092-9 2002 These results mean that CPT-11 and SN-38 interact with human P450 isoforms, such as CYP2A6, CYP2C9, and CYP3A4, in vitro and imply that the significant drug interactions involving CPT-11 may be caused by a mechanism-based inactivation of CYP3A4 by SN-38 as an active metabolite of CPT-11 rather than competitive inhibition. Irinotecan 180-186 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 238-244 11901092-9 2002 These results mean that CPT-11 and SN-38 interact with human P450 isoforms, such as CYP2A6, CYP2C9, and CYP3A4, in vitro and imply that the significant drug interactions involving CPT-11 may be caused by a mechanism-based inactivation of CYP3A4 by SN-38 as an active metabolite of CPT-11 rather than competitive inhibition. Irinotecan 248-253 cytochrome P450 family 2 subfamily A member 6 Homo sapiens 84-90 11901092-9 2002 These results mean that CPT-11 and SN-38 interact with human P450 isoforms, such as CYP2A6, CYP2C9, and CYP3A4, in vitro and imply that the significant drug interactions involving CPT-11 may be caused by a mechanism-based inactivation of CYP3A4 by SN-38 as an active metabolite of CPT-11 rather than competitive inhibition. Irinotecan 248-253 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 104-110 11901092-9 2002 These results mean that CPT-11 and SN-38 interact with human P450 isoforms, such as CYP2A6, CYP2C9, and CYP3A4, in vitro and imply that the significant drug interactions involving CPT-11 may be caused by a mechanism-based inactivation of CYP3A4 by SN-38 as an active metabolite of CPT-11 rather than competitive inhibition. Irinotecan 180-186 cytochrome P450 family 2 subfamily A member 6 Homo sapiens 84-90 11901092-9 2002 These results mean that CPT-11 and SN-38 interact with human P450 isoforms, such as CYP2A6, CYP2C9, and CYP3A4, in vitro and imply that the significant drug interactions involving CPT-11 may be caused by a mechanism-based inactivation of CYP3A4 by SN-38 as an active metabolite of CPT-11 rather than competitive inhibition. Irinotecan 180-186 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 92-98 11901092-9 2002 These results mean that CPT-11 and SN-38 interact with human P450 isoforms, such as CYP2A6, CYP2C9, and CYP3A4, in vitro and imply that the significant drug interactions involving CPT-11 may be caused by a mechanism-based inactivation of CYP3A4 by SN-38 as an active metabolite of CPT-11 rather than competitive inhibition. Irinotecan 180-186 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 104-110 11901092-9 2002 These results mean that CPT-11 and SN-38 interact with human P450 isoforms, such as CYP2A6, CYP2C9, and CYP3A4, in vitro and imply that the significant drug interactions involving CPT-11 may be caused by a mechanism-based inactivation of CYP3A4 by SN-38 as an active metabolite of CPT-11 rather than competitive inhibition. Irinotecan 180-186 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 238-244 11927002-1 2002 Breast cancer resistance protein (BCRP), an adenosine triphosphate-binding cassette transporter, confers resistance to a series of anticancer reagents, including mitoxantrone, SN-38 and topotecan. Irinotecan 176-181 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 0-32 11927002-1 2002 Breast cancer resistance protein (BCRP), an adenosine triphosphate-binding cassette transporter, confers resistance to a series of anticancer reagents, including mitoxantrone, SN-38 and topotecan. Irinotecan 176-181 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 34-38 11927002-2 2002 In the present study, we found that estrone and 17beta-estradiol potentiated the cytotoxicity of mitoxantrone, SN-38 and topotecan in BCRP-transduced K562 cells (K562 / BCRP). Irinotecan 111-116 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 134-138 11901092-1 2002 The inhibition and mechanism-based inactivation potencies of irinotecan (7-ethyl-10-[4-(1-piperidino)-1-piperidino]carbonyloxycamptothecin; CPT-11) and its active metabolite (7-ethyl-10-hydroxycamptothecin; SN-38) for human cytochrome P450 (P450) enzymes were investigated to evaluate the potential for drug interactions involving CPT-11 using microsomes from insect cells expressing specific human P450 isoforms. Irinotecan 73-138 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 224-245 11901092-3 2002 CPT-11 and SN-38 competitively inhibited CYP3A4 (testosterone 6 beta-hydroxylation) activity with K(i) values of 129 and 121 microM, respectively. Irinotecan 0-6 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 41-47 11901092-3 2002 CPT-11 and SN-38 competitively inhibited CYP3A4 (testosterone 6 beta-hydroxylation) activity with K(i) values of 129 and 121 microM, respectively. Irinotecan 11-16 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 41-47 11901092-4 2002 CYP2A6 (coumarin 7-hydroxylation) and CYP2C9 (diclofenac 4"-hydroxylation) activities exhibited a mixed type of inhibition comprising competitive and noncompetitive components in response to SN-38, the K(i) values being 181 and 156 microM, respectively. Irinotecan 191-196 cytochrome P450 family 2 subfamily A member 6 Homo sapiens 0-6 11901092-4 2002 CYP2A6 (coumarin 7-hydroxylation) and CYP2C9 (diclofenac 4"-hydroxylation) activities exhibited a mixed type of inhibition comprising competitive and noncompetitive components in response to SN-38, the K(i) values being 181 and 156 microM, respectively. Irinotecan 191-196 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 11894120-0 2002 Antisense anti-MDM2 mixed-backbone oligonucleotides enhance therapeutic efficacy of topoisomerase I inhibitor irinotecan in nude mice bearing human cancer xenografts: In vivo activity and mechanisms. Irinotecan 110-120 transformed mouse 3T3 cell double minute 2 Mus musculus 15-19 11894120-5 2002 In LS174T cells, the antisense oligonucleotide, but not the mismatch oligonucleotide, specifically inhibited MDM2 expression, resulting in a significant increase in irinotecan-associated p53 activation and p21 induction. Irinotecan 165-175 tumor protein p53 Homo sapiens 187-190 11894120-6 2002 In DLD-1 cells, the antisense oligonucleotide specifically inhibited MDM2 expression, resulting in a significant increase in irinotecan-associated p21 induction although mutant p53 levels remained unchanged. Irinotecan 125-135 MDM2 proto-oncogene Homo sapiens 69-73 11894120-6 2002 In DLD-1 cells, the antisense oligonucleotide specifically inhibited MDM2 expression, resulting in a significant increase in irinotecan-associated p21 induction although mutant p53 levels remained unchanged. Irinotecan 125-135 H3 histone pseudogene 16 Homo sapiens 147-150 11990699-2 2002 Recent studies have shown that irinotecan also undergoes oxidative metabolism by the P450 isozyme CYP3A4, leading to the formation of a minor inactive metabolite, 7-ethyl-10-[4-N-[(5-aminopentanoic acid)-1-piperidino]-carbonyloxy-camptothecin (APC). Irinotecan 31-41 APC regulator of WNT signaling pathway Homo sapiens 244-247 11990699-3 2002 The elucidation of this metabolic pathway suggests the potential for drug interactions when irinotecan is administered with other inducers or substrates of CYP3A4. Irinotecan 92-102 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 156-162 11805731-0 2002 Polymorphisms of UDP-glucuronosyltransferase and pharmacokinetics of irinotecan. Irinotecan 69-79 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 17-44 11805731-2 2002 SN-38 is further conjugated and detoxified by UDP-glucuronosyltransferase (UGT) to yield its beta-glucuronide (SN-38G). Irinotecan 0-5 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 46-73 11805731-2 2002 SN-38 is further conjugated and detoxified by UDP-glucuronosyltransferase (UGT) to yield its beta-glucuronide (SN-38G). Irinotecan 0-5 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 75-78 11805731-2 2002 SN-38 is further conjugated and detoxified by UDP-glucuronosyltransferase (UGT) to yield its beta-glucuronide (SN-38G). Irinotecan 111-116 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 46-73 11805731-2 2002 SN-38 is further conjugated and detoxified by UDP-glucuronosyltransferase (UGT) to yield its beta-glucuronide (SN-38G). Irinotecan 111-116 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 75-78 11805731-4 2002 Interindividual variation of sensitivity to irinotecan is related to large variations of biotransformation of the active metabolite SN-38, some of which would be caused by genetic polymorphism of UGT1A1, an isozyme responsible for the SN-38 glucuronidation. Irinotecan 44-54 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 196-202 11805731-4 2002 Interindividual variation of sensitivity to irinotecan is related to large variations of biotransformation of the active metabolite SN-38, some of which would be caused by genetic polymorphism of UGT1A1, an isozyme responsible for the SN-38 glucuronidation. Irinotecan 132-137 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 196-202 11805731-4 2002 Interindividual variation of sensitivity to irinotecan is related to large variations of biotransformation of the active metabolite SN-38, some of which would be caused by genetic polymorphism of UGT1A1, an isozyme responsible for the SN-38 glucuronidation. Irinotecan 235-240 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 196-202 11805731-9 2002 The 4 subjects carrying UGT1A1*28 allele had values of the AUC(SN-38)/AUC(SN-38G) above the 75th percentile of the total population, suggesting a potential pharmacogenetic/pharmacokinetic relationship. Irinotecan 63-68 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 24-30 11805731-9 2002 The 4 subjects carrying UGT1A1*28 allele had values of the AUC(SN-38)/AUC(SN-38G) above the 75th percentile of the total population, suggesting a potential pharmacogenetic/pharmacokinetic relationship. Irinotecan 74-79 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 24-30 11805731-11 2002 The large variation in the UGT activity being related to the genetic status would warrant pharmacogenetic-guided dosing of irinotecan. Irinotecan 123-133 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 27-30 12017271-2 2002 In this study we determined the therapeutic effects of an antisense anti-MDM2 oligonucleotide administered alone or in combination with the clinically used chemotherapeutic agents Paclitaxel and Irinotecan. Irinotecan 195-205 MDM2 proto-oncogene Homo sapiens 73-77 11700302-8 2002 The loss of ATR-dependent checkpoint function sensitized GM847 human fibroblasts to the cytotoxic effects of the topoisomerase I poisons TPT and 7-ethyl-10-hydroxycamptothecin, as assessed by inhibition of colony formation, increased trypan blue uptake, and development of apoptotic morphological changes. Irinotecan 145-175 ATR serine/threonine kinase Homo sapiens 12-15 15618702-1 2002 Three novel single nucleotide polymorphisms (SNPs) were found in the UDP-glucuronosyltransferase (UGT) 1A10 gene from 24 Japanese patients with various cancers who were administered the anti-tumor drug, irinotecan (CPT-11). Irinotecan 203-213 UDP glucuronosyltransferase family 1 member A10 Homo sapiens 69-107 15618649-2 2002 Since the biliary excretion of its active metabolite (SN-38) and SN-38 glucuronide (SN38-Glu), which are mediated by the multidrug resistance associated protein-2 (MRP2/ABCC2), has been proposed to be related to this gastrointestinal toxicity, we have attempted here to examine the potential of various therapeutic agents to interact with the biliary excretion in order to identify MRP2 inhibitors to prevent this toxicity. Irinotecan 54-59 ATP binding cassette subfamily C member 2 Homo sapiens 121-162 15618649-2 2002 Since the biliary excretion of its active metabolite (SN-38) and SN-38 glucuronide (SN38-Glu), which are mediated by the multidrug resistance associated protein-2 (MRP2/ABCC2), has been proposed to be related to this gastrointestinal toxicity, we have attempted here to examine the potential of various therapeutic agents to interact with the biliary excretion in order to identify MRP2 inhibitors to prevent this toxicity. Irinotecan 54-59 ATP binding cassette subfamily C member 2 Homo sapiens 164-168 15618661-3 2002 SN-38-selected PC-6/SN2-5H human lung carcinoma cells were shown to overexpress ABCG2 with the reduced intracellular accumulation of SN-38, the active metabolite of irinotecan. Irinotecan 0-5 proprotein convertase subtilisin/kexin type 5 Homo sapiens 15-19 15618661-3 2002 SN-38-selected PC-6/SN2-5H human lung carcinoma cells were shown to overexpress ABCG2 with the reduced intracellular accumulation of SN-38, the active metabolite of irinotecan. Irinotecan 0-5 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 80-85 15618661-3 2002 SN-38-selected PC-6/SN2-5H human lung carcinoma cells were shown to overexpress ABCG2 with the reduced intracellular accumulation of SN-38, the active metabolite of irinotecan. Irinotecan 133-138 proprotein convertase subtilisin/kexin type 5 Homo sapiens 15-19 15618661-3 2002 SN-38-selected PC-6/SN2-5H human lung carcinoma cells were shown to overexpress ABCG2 with the reduced intracellular accumulation of SN-38, the active metabolite of irinotecan. Irinotecan 133-138 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 80-85 15618649-2 2002 Since the biliary excretion of its active metabolite (SN-38) and SN-38 glucuronide (SN38-Glu), which are mediated by the multidrug resistance associated protein-2 (MRP2/ABCC2), has been proposed to be related to this gastrointestinal toxicity, we have attempted here to examine the potential of various therapeutic agents to interact with the biliary excretion in order to identify MRP2 inhibitors to prevent this toxicity. Irinotecan 54-59 ATP binding cassette subfamily C member 2 Homo sapiens 169-174 15618649-2 2002 Since the biliary excretion of its active metabolite (SN-38) and SN-38 glucuronide (SN38-Glu), which are mediated by the multidrug resistance associated protein-2 (MRP2/ABCC2), has been proposed to be related to this gastrointestinal toxicity, we have attempted here to examine the potential of various therapeutic agents to interact with the biliary excretion in order to identify MRP2 inhibitors to prevent this toxicity. Irinotecan 54-59 ATP binding cassette subfamily C member 2 Homo sapiens 382-386 15618661-3 2002 SN-38-selected PC-6/SN2-5H human lung carcinoma cells were shown to overexpress ABCG2 with the reduced intracellular accumulation of SN-38, the active metabolite of irinotecan. Irinotecan 165-175 proprotein convertase subtilisin/kexin type 5 Homo sapiens 15-19 15618702-1 2002 Three novel single nucleotide polymorphisms (SNPs) were found in the UDP-glucuronosyltransferase (UGT) 1A10 gene from 24 Japanese patients with various cancers who were administered the anti-tumor drug, irinotecan (CPT-11). Irinotecan 215-221 UDP glucuronosyltransferase family 1 member A10 Homo sapiens 69-107 15618661-3 2002 SN-38-selected PC-6/SN2-5H human lung carcinoma cells were shown to overexpress ABCG2 with the reduced intracellular accumulation of SN-38, the active metabolite of irinotecan. Irinotecan 165-175 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 80-85 15618661-4 2002 We have recently demonstrated that plasma membrane vesicles prepared from those cells transported SN-38 in an ATP-dependent manner, and it was suggested that ABCG2 is involved in the active extrusion of SN-38 from cancer cells. Irinotecan 98-103 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 158-163 15618661-4 2002 We have recently demonstrated that plasma membrane vesicles prepared from those cells transported SN-38 in an ATP-dependent manner, and it was suggested that ABCG2 is involved in the active extrusion of SN-38 from cancer cells. Irinotecan 203-208 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 158-163 15618661-8 2002 Contrary to our expectation, however, ATPase activity in the cell membranes expressing ABCG2 was stimulated by neither SN-38 nor rhodamine 123. Irinotecan 119-124 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 87-92 15618661-9 2002 It is suggested that there is a partner protein of ABCG2 required for heterodimer formation to exhibit transport activity toward SN-38. Irinotecan 129-134 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 51-56 11802813-2 2002 We found that a DNA repair protein, O(6)-methylguanine-DNA methyltransferase (MGMT), participated in resistance to irinotecan hydrochloride (CPT-11), its active metabolite SN-38, and a novel CPT derivative, DX-8951f. Irinotecan 115-139 O-6-methylguanine-DNA methyltransferase Homo sapiens 36-76 11773147-0 2002 Irinotecan dosing: does the CPT in CPT-11 stand for "Can"t Predict Toxicity"? Irinotecan 0-10 choline phosphotransferase 1 Homo sapiens 28-31 11773147-0 2002 Irinotecan dosing: does the CPT in CPT-11 stand for "Can"t Predict Toxicity"? Irinotecan 0-10 choline phosphotransferase 1 Homo sapiens 35-38 11802813-2 2002 We found that a DNA repair protein, O(6)-methylguanine-DNA methyltransferase (MGMT), participated in resistance to irinotecan hydrochloride (CPT-11), its active metabolite SN-38, and a novel CPT derivative, DX-8951f. Irinotecan 115-139 O-6-methylguanine-DNA methyltransferase Homo sapiens 78-82 11802813-2 2002 We found that a DNA repair protein, O(6)-methylguanine-DNA methyltransferase (MGMT), participated in resistance to irinotecan hydrochloride (CPT-11), its active metabolite SN-38, and a novel CPT derivative, DX-8951f. Irinotecan 141-147 O-6-methylguanine-DNA methyltransferase Homo sapiens 36-76 11802813-2 2002 We found that a DNA repair protein, O(6)-methylguanine-DNA methyltransferase (MGMT), participated in resistance to irinotecan hydrochloride (CPT-11), its active metabolite SN-38, and a novel CPT derivative, DX-8951f. Irinotecan 141-147 O-6-methylguanine-DNA methyltransferase Homo sapiens 78-82 11802813-2 2002 We found that a DNA repair protein, O(6)-methylguanine-DNA methyltransferase (MGMT), participated in resistance to irinotecan hydrochloride (CPT-11), its active metabolite SN-38, and a novel CPT derivative, DX-8951f. Irinotecan 172-177 O-6-methylguanine-DNA methyltransferase Homo sapiens 36-76 11802813-2 2002 We found that a DNA repair protein, O(6)-methylguanine-DNA methyltransferase (MGMT), participated in resistance to irinotecan hydrochloride (CPT-11), its active metabolite SN-38, and a novel CPT derivative, DX-8951f. Irinotecan 172-177 O-6-methylguanine-DNA methyltransferase Homo sapiens 78-82 12541026-0 2002 Irinotecan plus low-dose cisplatin for alpha-fetoprotein-producing gastric carcinoma with multiple liver metastases: report of two cases. Irinotecan 0-10 alpha fetoprotein Homo sapiens 39-56 11990381-2 2002 We have previously demonstrated the role of UGT1A1 enzyme in the glucuronidation of SN-38 and a significant correlation between in vitro glucuronidation of SN-38 and UGT1A1 gene promoter polymorphism. Irinotecan 156-161 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 166-172 11990381-4 2002 Here we report the results from a prospective clinical pharmacogenetic study to determine the significance of UGT1A1*28 polymorphism on irinotecan disposition and toxicity in patients with cancer. Irinotecan 136-146 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 110-116 11990381-10 2002 The results suggest that screening for UGT1A1*28 polymorphism may identify patients with lower SN-38 glucuronidation rates and greater susceptibility to irinotecan induced gastrointestinal and bone marrow toxicity. Irinotecan 95-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 39-45 11990381-10 2002 The results suggest that screening for UGT1A1*28 polymorphism may identify patients with lower SN-38 glucuronidation rates and greater susceptibility to irinotecan induced gastrointestinal and bone marrow toxicity. Irinotecan 153-163 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 39-45 11990381-0 2002 UGT1A1*28 polymorphism as a determinant of irinotecan disposition and toxicity. Irinotecan 43-53 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 11990381-2 2002 We have previously demonstrated the role of UGT1A1 enzyme in the glucuronidation of SN-38 and a significant correlation between in vitro glucuronidation of SN-38 and UGT1A1 gene promoter polymorphism. Irinotecan 84-89 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 44-50 11990381-2 2002 We have previously demonstrated the role of UGT1A1 enzyme in the glucuronidation of SN-38 and a significant correlation between in vitro glucuronidation of SN-38 and UGT1A1 gene promoter polymorphism. Irinotecan 84-89 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 166-172 11990381-2 2002 We have previously demonstrated the role of UGT1A1 enzyme in the glucuronidation of SN-38 and a significant correlation between in vitro glucuronidation of SN-38 and UGT1A1 gene promoter polymorphism. Irinotecan 156-161 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 44-50 11688982-1 2001 Overexpression of breast cancer resistance protein (BCRP) ABCG2 reportedly confers cancer cell resistance to camptothecin-based anticancer drugs, such as topotecan and 7-ethyl-10-hydroxycamptothecin (SN-38: the active metabolite of irinotecan). Irinotecan 168-198 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 18-50 11751522-4 2001 Exposure of parental Hct116 cells to clinically achievable concentrations of SN-38 (the active metabolite of CPT-11) induces p21 and a G(2) arrest. Irinotecan 77-82 H3 histone pseudogene 16 Homo sapiens 125-128 11751522-4 2001 Exposure of parental Hct116 cells to clinically achievable concentrations of SN-38 (the active metabolite of CPT-11) induces p21 and a G(2) arrest. Irinotecan 109-115 H3 histone pseudogene 16 Homo sapiens 125-128 11751522-6 2001 In contrast, Hct116 cells that are p21 deficient (p21-/- Hct116) readily undergo apoptosis after treatment with SN-38. Irinotecan 112-117 H3 histone pseudogene 16 Homo sapiens 35-38 11751522-6 2001 In contrast, Hct116 cells that are p21 deficient (p21-/- Hct116) readily undergo apoptosis after treatment with SN-38. Irinotecan 112-117 H3 histone pseudogene 16 Homo sapiens 50-53 11751522-10 2001 Sequential treatment with SN-38 followed by flavopiridol was associated with higher activation of caspase-3 and greater cleavage of both p21 and XIAP, an inhibitor of apoptosis, compared with other treatment schedules. Irinotecan 26-31 caspase 3 Homo sapiens 98-107 11751522-10 2001 Sequential treatment with SN-38 followed by flavopiridol was associated with higher activation of caspase-3 and greater cleavage of both p21 and XIAP, an inhibitor of apoptosis, compared with other treatment schedules. Irinotecan 26-31 H3 histone pseudogene 16 Homo sapiens 137-140 11751522-10 2001 Sequential treatment with SN-38 followed by flavopiridol was associated with higher activation of caspase-3 and greater cleavage of both p21 and XIAP, an inhibitor of apoptosis, compared with other treatment schedules. Irinotecan 26-31 X-linked inhibitor of apoptosis Homo sapiens 145-149 11742701-1 2001 In the last years, the main topoisomerase I inhibitors (TP1-I) (i.e. topotecan and irinotecan) have been used in combination chemotherapy in non-small cell lung cancer. Irinotecan 83-93 transition protein 1 Homo sapiens 56-59 11740913-10 2001 The in vitro activity of human erythrocyte acetylcholinesterase was significantly inhibited by irinotecan (-21.5% at 100 microM) or physostigmine (-84.8% at 1 microM), whereas SN-38 or camptothecin had no effect. Irinotecan 95-105 acetylcholinesterase Rattus norvegicus 43-63 11740913-10 2001 The in vitro activity of human erythrocyte acetylcholinesterase was significantly inhibited by irinotecan (-21.5% at 100 microM) or physostigmine (-84.8% at 1 microM), whereas SN-38 or camptothecin had no effect. Irinotecan 176-181 acetylcholinesterase Rattus norvegicus 43-63 11740913-11 2001 Rat atrial acetylcholinesterase was also significantly inhibited in vitro by irinotecan (-16.9% at 100 microM). Irinotecan 77-87 acetylcholinesterase Rattus norvegicus 11-31 11740913-13 2001 A direct activation of cholinergic receptors or an interaction with central nervous sites does not appear to account for these inhibitory actions, whereas a blockade of acetylcholinesterase seems to occur at concentrations of irinotecan that may not be relevant in clinical settings. Irinotecan 226-236 acetylcholinesterase Rattus norvegicus 169-189 11688982-0 2001 Transport of 7-ethyl-10-hydroxycamptothecin (SN-38) by breast cancer resistance protein ABCG2 in human lung cancer cells. Irinotecan 13-43 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 55-87 11688982-0 2001 Transport of 7-ethyl-10-hydroxycamptothecin (SN-38) by breast cancer resistance protein ABCG2 in human lung cancer cells. Irinotecan 13-43 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 88-93 11688982-0 2001 Transport of 7-ethyl-10-hydroxycamptothecin (SN-38) by breast cancer resistance protein ABCG2 in human lung cancer cells. Irinotecan 45-50 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 55-87 11688982-0 2001 Transport of 7-ethyl-10-hydroxycamptothecin (SN-38) by breast cancer resistance protein ABCG2 in human lung cancer cells. Irinotecan 45-50 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 88-93 11688982-1 2001 Overexpression of breast cancer resistance protein (BCRP) ABCG2 reportedly confers cancer cell resistance to camptothecin-based anticancer drugs, such as topotecan and 7-ethyl-10-hydroxycamptothecin (SN-38: the active metabolite of irinotecan). Irinotecan 168-198 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 52-56 11688982-1 2001 Overexpression of breast cancer resistance protein (BCRP) ABCG2 reportedly confers cancer cell resistance to camptothecin-based anticancer drugs, such as topotecan and 7-ethyl-10-hydroxycamptothecin (SN-38: the active metabolite of irinotecan). Irinotecan 168-198 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 58-63 11688982-1 2001 Overexpression of breast cancer resistance protein (BCRP) ABCG2 reportedly confers cancer cell resistance to camptothecin-based anticancer drugs, such as topotecan and 7-ethyl-10-hydroxycamptothecin (SN-38: the active metabolite of irinotecan). Irinotecan 200-205 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 18-50 11688982-1 2001 Overexpression of breast cancer resistance protein (BCRP) ABCG2 reportedly confers cancer cell resistance to camptothecin-based anticancer drugs, such as topotecan and 7-ethyl-10-hydroxycamptothecin (SN-38: the active metabolite of irinotecan). Irinotecan 200-205 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 52-56 11688982-1 2001 Overexpression of breast cancer resistance protein (BCRP) ABCG2 reportedly confers cancer cell resistance to camptothecin-based anticancer drugs, such as topotecan and 7-ethyl-10-hydroxycamptothecin (SN-38: the active metabolite of irinotecan). Irinotecan 200-205 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 58-63 11688982-1 2001 Overexpression of breast cancer resistance protein (BCRP) ABCG2 reportedly confers cancer cell resistance to camptothecin-based anticancer drugs, such as topotecan and 7-ethyl-10-hydroxycamptothecin (SN-38: the active metabolite of irinotecan). Irinotecan 232-242 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 18-50 11688982-1 2001 Overexpression of breast cancer resistance protein (BCRP) ABCG2 reportedly confers cancer cell resistance to camptothecin-based anticancer drugs, such as topotecan and 7-ethyl-10-hydroxycamptothecin (SN-38: the active metabolite of irinotecan). Irinotecan 232-242 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 52-56 11688982-1 2001 Overexpression of breast cancer resistance protein (BCRP) ABCG2 reportedly confers cancer cell resistance to camptothecin-based anticancer drugs, such as topotecan and 7-ethyl-10-hydroxycamptothecin (SN-38: the active metabolite of irinotecan). Irinotecan 232-242 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 58-63 11688982-2 2001 We have recently shown that SN-38-selected PC-6/SN2-5H human lung carcinoma cells overexpressed BCRP with the reduced intracellular accumulation of SN-38 and SN-38-glucuronide (S. Kawabata et al., Biochem. Irinotecan 28-33 proprotein convertase subtilisin/kexin type 5 Homo sapiens 43-47 11688982-2 2001 We have recently shown that SN-38-selected PC-6/SN2-5H human lung carcinoma cells overexpressed BCRP with the reduced intracellular accumulation of SN-38 and SN-38-glucuronide (S. Kawabata et al., Biochem. Irinotecan 28-33 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 96-100 11688982-2 2001 We have recently shown that SN-38-selected PC-6/SN2-5H human lung carcinoma cells overexpressed BCRP with the reduced intracellular accumulation of SN-38 and SN-38-glucuronide (S. Kawabata et al., Biochem. Irinotecan 148-153 proprotein convertase subtilisin/kexin type 5 Homo sapiens 43-47 11688982-2 2001 We have recently shown that SN-38-selected PC-6/SN2-5H human lung carcinoma cells overexpressed BCRP with the reduced intracellular accumulation of SN-38 and SN-38-glucuronide (S. Kawabata et al., Biochem. Irinotecan 148-153 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 96-100 11688982-7 2001 In the present study, we have examined whether BCRP transports SN-38 and/or SN-38-glucuronide in vitro, by using plasma membrane vesicles from the parental PC-6 and resistant PC-6/SN2-5H cells, where SN-38 and SN-38-glucuronide accumulation in membrane vesicles was measured by HPLC. Irinotecan 63-68 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 47-51 11688982-8 2001 Both SN-38 and SN-38-glucuronide were ATP-dependently transported into membrane vesicles prepared from PC-6/SN2-5H cells, whereas no transport activity was observed in membrane vesicles from PC-6 cells. Irinotecan 5-10 proprotein convertase subtilisin/kexin type 5 Homo sapiens 103-107 11688982-9 2001 The kinetic parameters of the transport observed in PC-6/SN2-5H vesicles were K(m) = 4.0 microM, V(max) = 714 pmol/mg/min for SN-38 and K(m) = 26 microM, V(max) = 833 pmol/mg/min for SN-38-glucuronide. Irinotecan 126-131 proprotein convertase subtilisin/kexin type 5 Homo sapiens 52-56 11688982-10 2001 These findings suggest that BCRP expressed in PC-6/SN2-5H cells transports both SN-38 and SN-38-glucuronide with a higher affinity toward SN-38. Irinotecan 80-85 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 28-32 11688982-10 2001 These findings suggest that BCRP expressed in PC-6/SN2-5H cells transports both SN-38 and SN-38-glucuronide with a higher affinity toward SN-38. Irinotecan 80-85 proprotein convertase subtilisin/kexin type 5 Homo sapiens 46-50 11688982-10 2001 These findings suggest that BCRP expressed in PC-6/SN2-5H cells transports both SN-38 and SN-38-glucuronide with a higher affinity toward SN-38. Irinotecan 90-95 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 28-32 11688982-10 2001 These findings suggest that BCRP expressed in PC-6/SN2-5H cells transports both SN-38 and SN-38-glucuronide with a higher affinity toward SN-38. Irinotecan 90-95 proprotein convertase subtilisin/kexin type 5 Homo sapiens 46-50 11822765-6 2001 When G-CSF was added, dose escalation beyond 350 mg/m2 could not be achieved due to grade 2-3 toxicities that prevented on-time retreatment with CPT-11. Irinotecan 145-151 colony stimulating factor 3 Homo sapiens 5-10 11691793-3 2001 We have studied the effects on NFkappaB activation of two topoisomerase poisons and DNA damaging agents that are used in chemotherapy: SN38 (7-ethyl-10-hydroxycamptothecin), the active metabolite of CPT11, and doxorubicin. Irinotecan 135-139 nuclear factor kappa B subunit 1 Homo sapiens 31-39 11691793-3 2001 We have studied the effects on NFkappaB activation of two topoisomerase poisons and DNA damaging agents that are used in chemotherapy: SN38 (7-ethyl-10-hydroxycamptothecin), the active metabolite of CPT11, and doxorubicin. Irinotecan 141-171 nuclear factor kappa B subunit 1 Homo sapiens 31-39 11602529-10 2001 These findings indicate that this newly detected metabolite is a CYP3A4-generated product that may be produced in hepatic microsomes of patients treated with CPT-11. Irinotecan 158-164 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 65-71 11716702-0 2001 Identification and activities of human carboxylesterases for the activation of CPT-11, a clinically approved anticancer drug. Irinotecan 79-85 carboxylesterase 1 Homo sapiens 39-56 11716702-7 2001 hCE-2 was 26-fold more active than human carboxylesterase 1 and was 65% as active as rabbit liver carboxylesterase, the most active CPT-11 hydrolyzing enzyme known. Irinotecan 132-138 carboxylesterase 2 Homo sapiens 0-5 11716702-7 2001 hCE-2 was 26-fold more active than human carboxylesterase 1 and was 65% as active as rabbit liver carboxylesterase, the most active CPT-11 hydrolyzing enzyme known. Irinotecan 132-138 liver carboxylesterase 1 Oryctolagus cuniculus 92-114 11716702-8 2001 The anti-p97 mAb 96.5 was linked to hCE-2, forming a conjugate that could bind to antigen-positive cancer cells and convert CPT-11 to SN-38. Irinotecan 124-130 carboxylesterase 2 Homo sapiens 36-41 11716702-8 2001 The anti-p97 mAb 96.5 was linked to hCE-2, forming a conjugate that could bind to antigen-positive cancer cells and convert CPT-11 to SN-38. Irinotecan 134-139 carboxylesterase 2 Homo sapiens 36-41 11761438-5 2001 Pretreatment of the SW480 primary colon carcinoma cell line with IFN-gamma, 5-FU, CPT-11 or CDDP enhanced ICAM-1 and Fas expression, resulting in Ag-specific CTL-mediated lysis involving Fas-dependent and -independent mechanisms. Irinotecan 82-88 intercellular adhesion molecule 1 Homo sapiens 106-112 11761438-6 2001 In contrast, pretreatment of the SW620 metastatic isolate, derived from the same patient, with IFN-gamma, CPT-11 or CDDP, but not 5-FU, enhanced ICAM-1 expression, resulting in Ag-specific CTL-mediated lysis via Fas-independent mechanisms only. Irinotecan 106-112 intercellular adhesion molecule 1 Homo sapiens 145-151 11705873-9 2001 Moreover, CPT-11, cisplatin, oxaliplatin, and Taxol remain highly cytotoxic in cells that overexpress TS. Irinotecan 10-16 thymidylate synthetase Homo sapiens 102-104 11602529-0 2001 A new metabolite of irinotecan in which formation is mediated by human hepatic cytochrome P-450 3A4. Irinotecan 20-30 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 79-99 11602529-3 2001 Several oxidative metabolites of CPT-11 have been identified in humans, including 7-ethyl-10-[4-N-(5-aminopentanoic acid)-1-piperidino]carbonyloxycamptothecin (APC) and 7-ethyl-10-(4-amino-1-piperidino)carbonyloxycamptothecin (NPC), generated by cytochrome P-450 3A4 (CYP3A4). Irinotecan 33-39 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 246-266 11602529-3 2001 Several oxidative metabolites of CPT-11 have been identified in humans, including 7-ethyl-10-[4-N-(5-aminopentanoic acid)-1-piperidino]carbonyloxycamptothecin (APC) and 7-ethyl-10-(4-amino-1-piperidino)carbonyloxycamptothecin (NPC), generated by cytochrome P-450 3A4 (CYP3A4). Irinotecan 33-39 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 268-274 11822765-11 2001 CONCLUSIONS: When administered every two weeks, the recommended phase II starting dose of CPT-11 is 250 mg/m2 when given alone and 300 mg/m2 when supported by G-CSF. Irinotecan 90-96 colony stimulating factor 3 Homo sapiens 159-164 11593056-7 2001 Irinotecan induced apoptosis with an increase of poly(ADP-ribose) polymerase (PARP) cleavage in SW-620 cells (60 versus 7% basal level). Irinotecan 0-10 poly(ADP-ribose) polymerase 1 Homo sapiens 49-76 11714450-9 2001 When we assessed the mRNA levels of the multidrug resistance-associated protein (MRP), which may be an efflux pump for DDP, the combination of paclitaxel or SN-38 with DDP down-regulated these levels, which are up-regulated by DDP alone. Irinotecan 157-162 ATP binding cassette subfamily C member 3 Homo sapiens 40-79 11714450-9 2001 When we assessed the mRNA levels of the multidrug resistance-associated protein (MRP), which may be an efflux pump for DDP, the combination of paclitaxel or SN-38 with DDP down-regulated these levels, which are up-regulated by DDP alone. Irinotecan 157-162 ATP binding cassette subfamily C member 3 Homo sapiens 81-84 11593056-7 2001 Irinotecan induced apoptosis with an increase of poly(ADP-ribose) polymerase (PARP) cleavage in SW-620 cells (60 versus 7% basal level). Irinotecan 0-10 poly(ADP-ribose) polymerase 1 Homo sapiens 78-82 11593056-8 2001 Pretreatment of these cells with oxaliplatin abolished the increase in PARP cleavage induced by irinotecan (29%). Irinotecan 96-106 poly(ADP-ribose) polymerase 1 Homo sapiens 71-75 11507714-8 2001 The intra- and inter-day precision of enzyme assay for UDP-glucuronosyltransferase (UGT) activity toward SN-38 in human liver microsomes was less than 4%. Irinotecan 105-110 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 55-82 12049482-0 2001 The inhibition of acetylcholinesterase by irinotecan and related camptothecins: key structural properties and experimental variables. Irinotecan 42-52 acetylcholinesterase (Cartwright blood group) Homo sapiens 18-38 11695848-0 2001 Human liver UDP-glucuronosyltransferase isoforms involved in the glucuronidation of 7-ethyl-10-hydroxycamptothecin. Irinotecan 84-114 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 12-39 11695848-2 2001 The human liver UDP-glucuronosyltransferase (UGT) isoforms involved in the glucuronidation of 7-ethyl-10-hydroxycamptothecin (SN-38), the active metabolite of irinotecan (CPT-11), have been studied using microsomes from human liver and insect cells expressing human UGTs (1A1, 1A3, 1A4, 1A6, 1A9, 2B7, 2B15). Irinotecan 94-124 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 16-43 11695848-2 2001 The human liver UDP-glucuronosyltransferase (UGT) isoforms involved in the glucuronidation of 7-ethyl-10-hydroxycamptothecin (SN-38), the active metabolite of irinotecan (CPT-11), have been studied using microsomes from human liver and insect cells expressing human UGTs (1A1, 1A3, 1A4, 1A6, 1A9, 2B7, 2B15). Irinotecan 94-124 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 45-48 11695848-2 2001 The human liver UDP-glucuronosyltransferase (UGT) isoforms involved in the glucuronidation of 7-ethyl-10-hydroxycamptothecin (SN-38), the active metabolite of irinotecan (CPT-11), have been studied using microsomes from human liver and insect cells expressing human UGTs (1A1, 1A3, 1A4, 1A6, 1A9, 2B7, 2B15). Irinotecan 126-131 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 16-43 11695848-2 2001 The human liver UDP-glucuronosyltransferase (UGT) isoforms involved in the glucuronidation of 7-ethyl-10-hydroxycamptothecin (SN-38), the active metabolite of irinotecan (CPT-11), have been studied using microsomes from human liver and insect cells expressing human UGTs (1A1, 1A3, 1A4, 1A6, 1A9, 2B7, 2B15). Irinotecan 126-131 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 45-48 11695848-2 2001 The human liver UDP-glucuronosyltransferase (UGT) isoforms involved in the glucuronidation of 7-ethyl-10-hydroxycamptothecin (SN-38), the active metabolite of irinotecan (CPT-11), have been studied using microsomes from human liver and insect cells expressing human UGTs (1A1, 1A3, 1A4, 1A6, 1A9, 2B7, 2B15). Irinotecan 159-169 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 16-43 11695848-2 2001 The human liver UDP-glucuronosyltransferase (UGT) isoforms involved in the glucuronidation of 7-ethyl-10-hydroxycamptothecin (SN-38), the active metabolite of irinotecan (CPT-11), have been studied using microsomes from human liver and insect cells expressing human UGTs (1A1, 1A3, 1A4, 1A6, 1A9, 2B7, 2B15). Irinotecan 159-169 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 45-48 11695848-2 2001 The human liver UDP-glucuronosyltransferase (UGT) isoforms involved in the glucuronidation of 7-ethyl-10-hydroxycamptothecin (SN-38), the active metabolite of irinotecan (CPT-11), have been studied using microsomes from human liver and insect cells expressing human UGTs (1A1, 1A3, 1A4, 1A6, 1A9, 2B7, 2B15). Irinotecan 171-177 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 16-43 11695848-2 2001 The human liver UDP-glucuronosyltransferase (UGT) isoforms involved in the glucuronidation of 7-ethyl-10-hydroxycamptothecin (SN-38), the active metabolite of irinotecan (CPT-11), have been studied using microsomes from human liver and insect cells expressing human UGTs (1A1, 1A3, 1A4, 1A6, 1A9, 2B7, 2B15). Irinotecan 171-177 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 45-48 11695848-4 2001 The glucuronidation of SN-38 was catalysed by UGT1A1, UGT1A3, UGT1A6 and UGT1A9 as well as by liver microsomes. Irinotecan 23-28 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 46-52 11695848-4 2001 The glucuronidation of SN-38 was catalysed by UGT1A1, UGT1A3, UGT1A6 and UGT1A9 as well as by liver microsomes. Irinotecan 23-28 UDP glucuronosyltransferase family 1 member A3 Homo sapiens 54-60 11695848-4 2001 The glucuronidation of SN-38 was catalysed by UGT1A1, UGT1A3, UGT1A6 and UGT1A9 as well as by liver microsomes. Irinotecan 23-28 UDP glucuronosyltransferase family 1 member A6 Homo sapiens 62-68 11695848-4 2001 The glucuronidation of SN-38 was catalysed by UGT1A1, UGT1A3, UGT1A6 and UGT1A9 as well as by liver microsomes. Irinotecan 23-28 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 73-79 11695848-5 2001 Among these UGT isoforms, UGT1A1 showed the highest activity of SN-38 glucuronidation at both low (1 microM) and high (200 microM) substrate concentrations. Irinotecan 64-69 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 12-15 11695848-5 2001 Among these UGT isoforms, UGT1A1 showed the highest activity of SN-38 glucuronidation at both low (1 microM) and high (200 microM) substrate concentrations. Irinotecan 64-69 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 26-32 11695848-10 2001 The UGT isoforms involved in SN-38 glucuronidation could be classified into two types: low-Km types such as UGT1A1 and UGT1A9, and high-Km types such as UGT1A3 and UGT1A6, in terms of affinity toward substrate. Irinotecan 29-34 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 4-7 11695848-10 2001 The UGT isoforms involved in SN-38 glucuronidation could be classified into two types: low-Km types such as UGT1A1 and UGT1A9, and high-Km types such as UGT1A3 and UGT1A6, in terms of affinity toward substrate. Irinotecan 29-34 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 108-114 11695848-10 2001 The UGT isoforms involved in SN-38 glucuronidation could be classified into two types: low-Km types such as UGT1A1 and UGT1A9, and high-Km types such as UGT1A3 and UGT1A6, in terms of affinity toward substrate. Irinotecan 29-34 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 119-125 11695848-10 2001 The UGT isoforms involved in SN-38 glucuronidation could be classified into two types: low-Km types such as UGT1A1 and UGT1A9, and high-Km types such as UGT1A3 and UGT1A6, in terms of affinity toward substrate. Irinotecan 29-34 UDP glucuronosyltransferase family 1 member A3 Homo sapiens 153-159 11695848-10 2001 The UGT isoforms involved in SN-38 glucuronidation could be classified into two types: low-Km types such as UGT1A1 and UGT1A9, and high-Km types such as UGT1A3 and UGT1A6, in terms of affinity toward substrate. Irinotecan 29-34 UDP glucuronosyltransferase family 1 member A6 Homo sapiens 164-170 11695848-14 2001 The activity of SN-38 glucuronidation by liver microsomes and UGT1A1 was effectively inhibited by bilirubin. Irinotecan 16-21 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 62-68 11695848-16 2001 Although cholic acid derivatives strongly inhibited the activity of SN-38 glucuronidation by UGT1A3, the inhibition profile did not parallel that in liver microsomes. Irinotecan 68-73 UDP glucuronosyltransferase family 1 member A3 Homo sapiens 93-99 11695848-18 2001 These results demonstrate that at least four UGT1A isoforms are responsible for SN-38 glucuronidation in human livers, and suggest that the role and contribution of each differ substantially. Irinotecan 80-85 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 45-50 11559526-6 2001 Cross-resistance studies suggest that, compared with wild-type MXR/BCRP/ABCP, cells having an R482T mutation have higher anthracycline resistance, whereas an R482G mutation seems to confer relatively less resistance to SN-38 and topotecan. Irinotecan 219-224 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 63-66 11559526-6 2001 Cross-resistance studies suggest that, compared with wild-type MXR/BCRP/ABCP, cells having an R482T mutation have higher anthracycline resistance, whereas an R482G mutation seems to confer relatively less resistance to SN-38 and topotecan. Irinotecan 219-224 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 67-71 11559526-6 2001 Cross-resistance studies suggest that, compared with wild-type MXR/BCRP/ABCP, cells having an R482T mutation have higher anthracycline resistance, whereas an R482G mutation seems to confer relatively less resistance to SN-38 and topotecan. Irinotecan 219-224 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 72-76 11474256-8 2001 Maximum tolerated dosage (MTD) for the topotecan and irinotecan combination was defined as that which permitted 4 weeks of topotecan and irinotecan administration with G-CSF at or near the dose intensity reported for each single agent. Irinotecan 53-63 colony stimulating factor 2 Homo sapiens 168-173 12049482-2 2001 Recently, we demonstrated that CPT-11 (lactone) is also a potent inhibitor of human acetylcholinesterase (AChE) at clinically relevant concentrations. Irinotecan 31-37 acetylcholinesterase (Cartwright blood group) Homo sapiens 84-104 12049482-2 2001 Recently, we demonstrated that CPT-11 (lactone) is also a potent inhibitor of human acetylcholinesterase (AChE) at clinically relevant concentrations. Irinotecan 31-37 acetylcholinesterase (Cartwright blood group) Homo sapiens 106-110 12049482-5 2001 Those compounds possessing N-substitutions at C-10 were all found to inhibit AChE in a similar kinetic manner to CPT-11, but with a broad range of potencies. Irinotecan 113-119 acetylcholinesterase (Cartwright blood group) Homo sapiens 77-81 11507714-8 2001 The intra- and inter-day precision of enzyme assay for UDP-glucuronosyltransferase (UGT) activity toward SN-38 in human liver microsomes was less than 4%. Irinotecan 105-110 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 84-87 11376565-0 2001 CPT-11 alters the circadian rhythm of dihydropyrimidine dehydrogenase mRNA in mouse liver. Irinotecan 0-6 dihydropyrimidine dehydrogenase Mus musculus 38-69 11455023-2 2001 We identified a rabbit liver carboxylesterase (CE) that was very efficient at CPT-11 metabolism; however, a human homolog that was more than 81% identical to this protein activated the drug poorly. Irinotecan 78-84 liver carboxylesterase 1 Oryctolagus cuniculus 23-45 11724314-0 2001 Streptococcal preparation OK-432 enhances the antitumor activity of CPT-11 by increasing Th1-cytokine production in mice. Irinotecan 68-74 negative elongation factor complex member C/D, Th1l Mus musculus 89-92 11724314-4 2001 RESULTS: SN-38, the active metabolite of CPT-11, exerted dose-dependent inhibition of interferon (IFN)-gamma production induced by OK-432 in mouse splenocytes. Irinotecan 9-14 interferon gamma Mus musculus 86-108 11724314-4 2001 RESULTS: SN-38, the active metabolite of CPT-11, exerted dose-dependent inhibition of interferon (IFN)-gamma production induced by OK-432 in mouse splenocytes. Irinotecan 41-47 interferon gamma Mus musculus 86-108 11724314-5 2001 In contrast, the optimum concentration of SN-38 increased interleukin (IL)-6 and IL-12 production by OK-432-activated splenocytes. Irinotecan 42-47 interleukin 6 Mus musculus 58-76 11724314-7 2001 Investigation of cytokine production showed that CPT-11 treatment principally inhibited IL-12 and IFN-gamma production, which was improved by the combined administration with OK-432. Irinotecan 49-55 interferon gamma Mus musculus 98-107 11724314-8 2001 CONCLUSION: These results indicate that CPT-11 inhibits type-1 T helper (Th1) cells despite its potential to stimulate macrophages and that OK-432 enhances the antitumor activity of CPT-11 by increasing Th1-cytokine production. Irinotecan 40-46 negative elongation factor complex member C/D, Th1l Mus musculus 73-76 11724314-8 2001 CONCLUSION: These results indicate that CPT-11 inhibits type-1 T helper (Th1) cells despite its potential to stimulate macrophages and that OK-432 enhances the antitumor activity of CPT-11 by increasing Th1-cytokine production. Irinotecan 182-188 negative elongation factor complex member C/D, Th1l Mus musculus 203-206 11431344-0 2001 Sensitization of human tumor cells to CPT-11 via adenoviral-mediated delivery of a rabbit liver carboxylesterase. Irinotecan 38-44 liver carboxylesterase 1 Oryctolagus cuniculus 90-112 11351255-5 2001 Cotreatment of N-acetyl-Asp-Glu-Val-Asp aldehyde (AC-DEVD-CHO), a caspase 3 inhibitor prevented cytotoxic effect of VP-16 and SN-38 (p<0.01). Irinotecan 126-131 caspase 3 Homo sapiens 66-75 11419031-8 2001 Furthermore, a recent randomized trial showed that the combination of a newer agent (irinotecan) with cisplatin was superior to a standard etoposide and cisplatin regimen in patients with newly diagnosed extensive-stage SCLC. Irinotecan 85-95 SCLC1 Homo sapiens 220-224 11489791-5 2001 CPT-11 is subject to extensive metabolic conversion by various enzyme systems, including esterases to form SN-38, UGT1A1 mediating glucuronidation of SN-38, as well as CYP3A4, which forms several pharmacologically inactive oxidation products. Irinotecan 0-6 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 114-120 11489791-5 2001 CPT-11 is subject to extensive metabolic conversion by various enzyme systems, including esterases to form SN-38, UGT1A1 mediating glucuronidation of SN-38, as well as CYP3A4, which forms several pharmacologically inactive oxidation products. Irinotecan 150-155 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 114-120 11395570-4 2001 Both the lactone and carboxylate forms of CPT-11 and SN-38 were actively transported across the cell monolayers, mainly by the apical-localized P-gp pump. Irinotecan 42-48 ATP binding cassette subfamily B member 1 Homo sapiens 144-148 11395570-4 2001 Both the lactone and carboxylate forms of CPT-11 and SN-38 were actively transported across the cell monolayers, mainly by the apical-localized P-gp pump. Irinotecan 53-58 ATP binding cassette subfamily B member 1 Homo sapiens 144-148 11395570-8 2001 In contrast, SN-38 efficacy decreased in the presence of P-gp inhibitors due to active transport toward the basolateral side, thereby reducing drug accumulation. Irinotecan 13-18 ATP binding cassette subfamily B member 1 Homo sapiens 57-61 11350876-7 2001 Our findings indicate that bacterial beta-glucuronidase plays a crucial role in CPT-11-induced diarrhea without affecting enterocycling and systemic SN-38 levels. Irinotecan 80-86 glucuronidase beta Homo sapiens 37-55 11376565-3 2001 To optimize the schedule of the CPT-11 plus 5-FU combination, we investigated the effect of CPT-11 on the circadian rhythm of DPD in vivo. Irinotecan 92-98 dihydropyrimidine dehydrogenase Mus musculus 126-129 11376565-5 2001 After intravenous administration of CPT-11 (30 mg / kg) at 20:00, the circadian rhythm of the DPD mRNA level in the liver was no longer observed 18 h later (14:00), but it was unaffected 6 and 18 h later (at 14:00 and 02:00) when CPT-11 was given at 08:00. Irinotecan 36-42 dihydropyrimidine dehydrogenase Mus musculus 94-97 11376565-5 2001 After intravenous administration of CPT-11 (30 mg / kg) at 20:00, the circadian rhythm of the DPD mRNA level in the liver was no longer observed 18 h later (14:00), but it was unaffected 6 and 18 h later (at 14:00 and 02:00) when CPT-11 was given at 08:00. Irinotecan 230-236 dihydropyrimidine dehydrogenase Mus musculus 94-97 11376565-6 2001 In addition, a dose-dependent lengthening of the period of the circadian rhythm of DPD was observed for 42 h after intravenous injection of CPT-11 at 20:00. Irinotecan 140-146 dihydropyrimidine dehydrogenase Mus musculus 83-86 11376565-7 2001 The levels of DPD protein and activity at 21 h after administration of CPT-11 (at 17:00) were significantly higher than at 9 h (at 05:00). Irinotecan 71-77 dihydropyrimidine dehydrogenase Mus musculus 14-17 11376565-8 2001 These results suggest that CPT-11 may influence the circadian rhythm of DPD at the transcriptional level. Irinotecan 27-33 dihydropyrimidine dehydrogenase Mus musculus 72-75 11376565-9 2001 Modulation of the circadian rhythm of DPD by CPT-11 may be a factor in optimizing the combination of 5-FU and CPT-11. Irinotecan 45-51 dihydropyrimidine dehydrogenase Mus musculus 38-41 11376565-9 2001 Modulation of the circadian rhythm of DPD by CPT-11 may be a factor in optimizing the combination of 5-FU and CPT-11. Irinotecan 110-116 dihydropyrimidine dehydrogenase Mus musculus 38-41 11309308-16 2001 delivered BCRP substrates, e.g., topotecan or irinotecan, by using a BCRP inhibitor. Irinotecan 46-56 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 10-14 11309308-16 2001 delivered BCRP substrates, e.g., topotecan or irinotecan, by using a BCRP inhibitor. Irinotecan 46-56 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 69-73 11309344-7 2001 From the combined data, we conclude that the affinities of topoisomerase I drugs for BCRP are, in decreasing order: SN-38 > topotecan > 9-aminocamptothecin approximately CPT-11 > NX211 > DX8951f > BNP1350. Irinotecan 116-121 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 85-89 11309344-7 2001 From the combined data, we conclude that the affinities of topoisomerase I drugs for BCRP are, in decreasing order: SN-38 > topotecan > 9-aminocamptothecin approximately CPT-11 > NX211 > DX8951f > BNP1350. Irinotecan 176-182 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 85-89 11280753-3 2001 The RC0.1 and RC1 cells have high cross-resistance to CPT derivatives including SN-38 and topotecan, but are not cross-resistant to the non-top1 inhibitors etoposide, doxorubicin, and vincristine. Irinotecan 80-85 chromobox 8 Homo sapiens 14-17 11259359-1 2001 Amonafide and irinotecan are anticancer drugs representative of the clinical relevance of N-acetyltransferase (NAT) and uridine diphosphate glucuronosyltransferase (UGT) polymorphisms in cancer chemotherapy, respectively. Irinotecan 14-24 bromodomain containing 2 Homo sapiens 90-109 11259359-1 2001 Amonafide and irinotecan are anticancer drugs representative of the clinical relevance of N-acetyltransferase (NAT) and uridine diphosphate glucuronosyltransferase (UGT) polymorphisms in cancer chemotherapy, respectively. Irinotecan 14-24 bromodomain containing 2 Homo sapiens 111-114 11259359-1 2001 Amonafide and irinotecan are anticancer drugs representative of the clinical relevance of N-acetyltransferase (NAT) and uridine diphosphate glucuronosyltransferase (UGT) polymorphisms in cancer chemotherapy, respectively. Irinotecan 14-24 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 120-163 11259359-1 2001 Amonafide and irinotecan are anticancer drugs representative of the clinical relevance of N-acetyltransferase (NAT) and uridine diphosphate glucuronosyltransferase (UGT) polymorphisms in cancer chemotherapy, respectively. Irinotecan 14-24 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 165-168 11259359-7 2001 SN-38, the active metabolite of irinotecan, is glucuronidated to the inactive SN-38 glucuronide by UGT1A1, the isoform catalyzing bilirubin glucuronidation. Irinotecan 0-5 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 99-105 11259359-7 2001 SN-38, the active metabolite of irinotecan, is glucuronidated to the inactive SN-38 glucuronide by UGT1A1, the isoform catalyzing bilirubin glucuronidation. Irinotecan 32-42 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 99-105 11259359-13 2001 A phenotyping procedure for UGT1A1 has not been identified and genotyping of the UGT1A1 promoter in patients receiving irinotecan may identify patients at increased risk of toxicity. Irinotecan 119-129 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 81-87 11259359-14 2001 A clinical trial at the University of Chicago is ongoing to demonstrate the predictive significance of UGT1A1 genotyping for irinotecan pharmacodynamics. Irinotecan 125-135 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 103-109 11115510-3 2001 Inhibiting the degradation of IkappaBalpha by pretreatment with the proteasome inhibitor MG-132 significantly inhibited NF-kappaB activation and apoptosis but not DNA damage induced by SN-38 or VP-16. Irinotecan 185-190 NFKB inhibitor alpha Homo sapiens 30-42 11115510-4 2001 Transfection of NSCLC-3 or NSCLC-5 cells with dominant negative mutant IkappaBalpha (mIkappaBalpha) inhibited SN-38 or VP-16 induced transcription and DNA binding activity of NF-kappaB without altering drug-induced apoptosis. Irinotecan 110-115 NFKB inhibitor alpha Homo sapiens 71-83 11177741-4 2001 Preliminary data suggest that gene expression levels of topoisomerase I, p21, bcl-2, and ICE may be predictive of response to therapy with CPT-11. Irinotecan 139-145 H3 histone pseudogene 16 Homo sapiens 73-76 11265410-0 2001 [A case of AFP-producing gastric cancer responding to low-dose CPT-11 and low-dose cisplatin combination chemotherapy]. Irinotecan 63-69 alpha fetoprotein Homo sapiens 11-14 11177741-4 2001 Preliminary data suggest that gene expression levels of topoisomerase I, p21, bcl-2, and ICE may be predictive of response to therapy with CPT-11. Irinotecan 139-145 BCL2 apoptosis regulator Homo sapiens 78-83 11177741-4 2001 Preliminary data suggest that gene expression levels of topoisomerase I, p21, bcl-2, and ICE may be predictive of response to therapy with CPT-11. Irinotecan 139-145 carboxylesterase 2 Homo sapiens 89-92 11177741-5 2001 Increased toxicity seen in patients treated with CPT-11 may be explained by polymorphism in the UGT1A1 gene, which is responsible for glucuronidation of the active metabolite of CPT-11. Irinotecan 49-55 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 96-102 11177741-5 2001 Increased toxicity seen in patients treated with CPT-11 may be explained by polymorphism in the UGT1A1 gene, which is responsible for glucuronidation of the active metabolite of CPT-11. Irinotecan 178-184 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 96-102 11265410-2 2001 We report an AFP-producing gastric cancer that showed a partial response to low-dose CPT-11 and low-dose cisplatin combination chemotherapy. Irinotecan 85-91 alpha fetoprotein Homo sapiens 13-16 11265410-3 2001 AFP-producing gastric cancers successfully treated with chemotherapy have been reported, but to our knowledge this is the first report of successful treatment with low-dose CPT-11 and low-dose cisplatin combination chemotherapy. Irinotecan 173-179 alpha fetoprotein Homo sapiens 0-3 11162657-0 2001 Breast cancer resistance protein directly confers SN-38 resistance of lung cancer cells. Irinotecan 50-55 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 0-32 11230497-2 2001 With the recognition that topoisomerase-I (TOP-I) is an important therapeutic target in cancer therapy, irinotecan, a semisynthetic TOP-I-interactive camptothecin derivative, has been clinically established in the treatment of colorectal cancer. Irinotecan 104-114 DNA topoisomerase I Homo sapiens 26-41 11230497-2 2001 With the recognition that topoisomerase-I (TOP-I) is an important therapeutic target in cancer therapy, irinotecan, a semisynthetic TOP-I-interactive camptothecin derivative, has been clinically established in the treatment of colorectal cancer. Irinotecan 104-114 DNA topoisomerase I Homo sapiens 43-48 11230497-2 2001 With the recognition that topoisomerase-I (TOP-I) is an important therapeutic target in cancer therapy, irinotecan, a semisynthetic TOP-I-interactive camptothecin derivative, has been clinically established in the treatment of colorectal cancer. Irinotecan 104-114 DNA topoisomerase I Homo sapiens 132-137 11162657-2 2001 BCRP-overexpressing cells show cross-resistance to camptothecin derivatives such as irinotecan, SN-38 (the active metabolite of irinotecan), and topotecan. Irinotecan 84-94 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 0-4 11162657-2 2001 BCRP-overexpressing cells show cross-resistance to camptothecin derivatives such as irinotecan, SN-38 (the active metabolite of irinotecan), and topotecan. Irinotecan 96-101 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 0-4 11162657-2 2001 BCRP-overexpressing cells show cross-resistance to camptothecin derivatives such as irinotecan, SN-38 (the active metabolite of irinotecan), and topotecan. Irinotecan 128-138 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 0-4 11162657-5 2001 The intracellular SN-38 accumulation was reduced in the sublines, and BCRP mRNA was overexpressed in proportion to the degree of SN-38 resistance. Irinotecan 129-134 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 70-74 11162657-6 2001 These findings suggest that BCRP confers SN-38 resistance in the sublines. Irinotecan 41-46 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 28-32 11162657-9 2001 These data indicate that BCRP is directly involved with SN-38 resistance, by efflux transport of SN-38. Irinotecan 56-61 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 25-29 11162657-9 2001 These data indicate that BCRP is directly involved with SN-38 resistance, by efflux transport of SN-38. Irinotecan 97-102 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 25-29 11142689-0 2000 Phase I study of carboplatin, docetaxel and irinotecan with recombinant human granulocyte colony stimulating factor support in patients with advanced non-small cell lung cancer. Irinotecan 44-54 colony stimulating factor 3 Homo sapiens 78-115 11162602-4 2001 The reduction in cyclin B1 expression and G2 arrest were also seen after treatment with etoposide and irinotecan. Irinotecan 102-112 cyclin B1 Homo sapiens 17-26 11212269-4 2001 We report here that a human degradation-resistant GLP-2 analogue, h[Gly2]-GLP-2 significantly improves survival, reduces bacteremia, attenuates epithelial injury, and inhibits crypt apoptosis in the murine gastrointestinal tract after administration of topoisomerase I inhibitor irinotecan hydrochloride or the antimetabolite 5-fluorouracil. Irinotecan 279-303 glucagon Homo sapiens 74-79 11212277-0 2001 The HER tyrosine kinase inhibitor CI1033 enhances cytotoxicity of 7-ethyl-10-hydroxycamptothecin and topotecan by inhibiting breast cancer resistance protein-mediated drug efflux. Irinotecan 66-96 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 125-157 11212277-10 2001 Moreover, CI1033 enhanced the uptake and cytotoxicity of SN-38 and TPT in cells transfected with BCRP but not empty vector. Irinotecan 57-62 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 97-101 11783425-0 2001 UGT1A1 polymorphism predicts irinotecan toxicity: evolving proof. Irinotecan 29-39 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 11212269-4 2001 We report here that a human degradation-resistant GLP-2 analogue, h[Gly2]-GLP-2 significantly improves survival, reduces bacteremia, attenuates epithelial injury, and inhibits crypt apoptosis in the murine gastrointestinal tract after administration of topoisomerase I inhibitor irinotecan hydrochloride or the antimetabolite 5-fluorouracil. Irinotecan 279-303 glucagon Homo sapiens 50-55 11156391-2 2000 Irinotecan is metabolized to form active SN-38, which is further conjugated and detoxified by UDP-glucuronosyltransferase (UGT) 1A1 enzyme. Irinotecan 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 94-131 11156391-2 2000 Irinotecan is metabolized to form active SN-38, which is further conjugated and detoxified by UDP-glucuronosyltransferase (UGT) 1A1 enzyme. Irinotecan 41-46 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 94-131 11156391-3 2000 Genetic polymorphisms of the UGT1A1 would affect an interindividual variation of the toxicity by irinotecan via the alternation of bioavailability of SN-38. Irinotecan 97-107 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 29-35 11156391-3 2000 Genetic polymorphisms of the UGT1A1 would affect an interindividual variation of the toxicity by irinotecan via the alternation of bioavailability of SN-38. Irinotecan 150-155 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 29-35 11156391-4 2000 In this case-control study, retrospective review of clinical records and determination of UGT1A1 polymorphisms were performed to investigate whether a patient with the variant UGT1A1 genotypes would be at higher risk for severe toxicity by irinotecan. Irinotecan 240-250 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 176-182 11156391-9 2000 Multivariate analysis suggested that the genotype either heterozygous or homozygous for UGT1A1*28 would be a significant risk factor for severe toxicity by irinotecan (P < 0.001; odds ratio, 7.23; 95% confidence interval, 2.52-22.3). Irinotecan 156-166 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 88-94 11156391-13 2000 We suggest that determination of the UGT1A1 genotypes might be clinically useful for predicting severe toxicity by irinotecan in cancer patients. Irinotecan 115-125 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 37-43 11156391-14 2000 This research warrants a prospective trial to corroborate the usefulness of gene diagnosis of UGT1A1 polymorphisms prior tb irinotecan chemotherapy. Irinotecan 124-134 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 94-100 11137204-7 2000 RESULTS: At the dose of CPT-11 120 mg/m2, three out of four enrolled patients presented DLTs (grade 4 neutropenia, febrile neutropenia and delayed diarrhea); the addition of G-CSF at this level did not permit further dose-escalation. Irinotecan 24-30 colony stimulating factor 3 Homo sapiens 174-179 11142689-1 2000 A phase I study was conducted in patients with stage IIIB or IV non-small cell lung cancer to determine the maximum tolerated dose (MTD) of irinotecan combined with a fixed schedule of docetaxel and carboplatin with recombinant human granulocyte colony stimulating factor (rhG-CSF) (nartograstim) support. Irinotecan 140-150 colony stimulating factor 3 Homo sapiens 234-271 11084429-6 2000 We found a good correlation between inactivation of TGFB-RII, BAX or hMSH3 genes and tumor response to CPT-11 (P =0.002). Irinotecan 103-109 transforming growth factor beta 1 Homo sapiens 52-56 11108431-6 2000 With regard to irinotecan, patients with Gilbert"s syndrome phenotype have reduced inactivation of the active topoisomerase I inhibitor 7-ethyl-10-hydroxycamptothecin (SN-38) caused by a mutation in the UDP-glucuronosyltransferase 1A1 gene promoter. Irinotecan 15-25 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 203-234 11108431-6 2000 With regard to irinotecan, patients with Gilbert"s syndrome phenotype have reduced inactivation of the active topoisomerase I inhibitor 7-ethyl-10-hydroxycamptothecin (SN-38) caused by a mutation in the UDP-glucuronosyltransferase 1A1 gene promoter. Irinotecan 136-166 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 203-234 11108431-6 2000 With regard to irinotecan, patients with Gilbert"s syndrome phenotype have reduced inactivation of the active topoisomerase I inhibitor 7-ethyl-10-hydroxycamptothecin (SN-38) caused by a mutation in the UDP-glucuronosyltransferase 1A1 gene promoter. Irinotecan 168-173 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 203-234 11084429-6 2000 We found a good correlation between inactivation of TGFB-RII, BAX or hMSH3 genes and tumor response to CPT-11 (P =0.002). Irinotecan 103-109 BCL2 associated X, apoptosis regulator Homo sapiens 62-65 11084429-6 2000 We found a good correlation between inactivation of TGFB-RII, BAX or hMSH3 genes and tumor response to CPT-11 (P =0.002). Irinotecan 103-109 mutS homolog 3 Homo sapiens 69-74 10912951-4 2000 The etoposide-resistant line with the highest P-gp level was cross-resistant also to SN-38, CPT-11 and topotecan (TPT), but not to 9-aminocamptothecin (9-AC), CPT and DX-8951f. Irinotecan 85-90 ATP binding cassette subfamily B member 1 Homo sapiens 46-50 11081458-8 2000 A 48-year-old patient with a HCC that had developed within a normal liver but of very poor prognosis because of a multifocal primary tumor with a large nodule measuring 10 cm of diameter, associated with a portal thrombosis, could tolerate very intensive treatment with irinotecan using doses up to 700 mg/m2 every 2 weeks and was responsive to treatment as measured by alpha-fetoprotein levels. Irinotecan 270-280 alpha fetoprotein Homo sapiens 370-387 10999728-2 2000 In the liver, SN-38 is glucuronidated (SN-38G) by UGT1A1, which also conjugates bilirubin. Irinotecan 14-19 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 50-56 10999728-2 2000 In the liver, SN-38 is glucuronidated (SN-38G) by UGT1A1, which also conjugates bilirubin. Irinotecan 39-44 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 50-56 10999728-8 2000 Interestingly, individual levels of fecal beta-glucuronidase, which is known to mediate SN-38G hydrolysis, were not related to any of the SN-38 kinetic parameters (r = 0.09; P = 0.26), suggesting that interindividual variation in this enzyme is unimportant in explaining SN-38 pharmacokinetic variability. Irinotecan 88-93 glucuronidase beta Homo sapiens 42-60 10971167-1 2000 OBJECTIVE: This study was conducted in refractory or relapsed small-cell lung cancer to determine activity and toxicity of the combination of cisplatin, ifosfamide, and irinotecan with rhG-CSF support. Irinotecan 169-179 colony stimulating factor 2 Homo sapiens 189-192 10912947-7 2000 Sample pretreatment procedures of the CPT analogs topotecan, irinotecan, 9-aminocamptothecin and lurtotecan are summarized and discussed in this review. Irinotecan 61-71 choline phosphotransferase 1 Homo sapiens 38-41 10912951-4 2000 The etoposide-resistant line with the highest P-gp level was cross-resistant also to SN-38, CPT-11 and topotecan (TPT), but not to 9-aminocamptothecin (9-AC), CPT and DX-8951f. Irinotecan 92-98 ATP binding cassette subfamily B member 1 Homo sapiens 46-50 10811101-4 2000 We show that the topoisomerase I inhibitor 7-ethyl-10-[4-(1-piperidino)-1-piperidino]carbonyloxycamptothecin (CPT-11) activates NF-kappaB in most colorectal cancer cell lines. Irinotecan 110-116 nuclear factor kappa B subunit 1 Homo sapiens 128-137 10811101-4 2000 We show that the topoisomerase I inhibitor 7-ethyl-10-[4-(1-piperidino)-1-piperidino]carbonyloxycamptothecin (CPT-11) activates NF-kappaB in most colorectal cancer cell lines. Irinotecan 43-108 nuclear factor kappa B subunit 1 Homo sapiens 128-137 10811101-6 2000 These data demonstrate that the protection from apoptosis induced in response to CPT-11 treatment is effectively inhibited by the transient inhibition of NF-kappaB in a variety of human colon cancer cell lines and in a tumor xenograft model, resulting in a significantly enhanced tumoricidal response to CPT-11 via increased induction of apoptosis. Irinotecan 81-87 nuclear factor kappa B subunit 1 Homo sapiens 154-163 10811101-6 2000 These data demonstrate that the protection from apoptosis induced in response to CPT-11 treatment is effectively inhibited by the transient inhibition of NF-kappaB in a variety of human colon cancer cell lines and in a tumor xenograft model, resulting in a significantly enhanced tumoricidal response to CPT-11 via increased induction of apoptosis. Irinotecan 304-310 nuclear factor kappa B subunit 1 Homo sapiens 154-163 10738246-2 2000 We have found that ectopic p16(INK4) expression increased cellular sensitivity of human non-small-cell-lung-cancer (NSCLC) A549 cells to a selective growth-inhibitory effect induced by the topoisomerase-I inhibitor 11, 7-ethyl-10-[4-(1-piperidino)-1-piperidino] carbonyloxy camptothecin (CPT-11) in vitro. Irinotecan 288-294 cyclin dependent kinase inhibitor 2A Homo sapiens 31-35 10815927-0 2000 Metabolism of irinotecan (CPT-11) by CYP3A4 and CYP3A5 in humans. Irinotecan 14-24 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 37-43 10815927-0 2000 Metabolism of irinotecan (CPT-11) by CYP3A4 and CYP3A5 in humans. Irinotecan 14-24 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 48-54 10815927-0 2000 Metabolism of irinotecan (CPT-11) by CYP3A4 and CYP3A5 in humans. Irinotecan 26-32 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 37-43 10815927-0 2000 Metabolism of irinotecan (CPT-11) by CYP3A4 and CYP3A5 in humans. Irinotecan 26-32 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 48-54 10815927-2 2000 The objective of this study was to determine the oxidative metabolites of CPT-11 recovered in human urine samples and to identify cytochrome P450 (CYP) involved in their formation. Irinotecan 74-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 147-150 10815927-7 2000 Only CYP3A4 produced both APC and NPC, resulting from the oxidation of the piperidinylpiperidine side chain of CPT-11 along with metabolite M2. Irinotecan 111-117 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 5-11 10815927-8 2000 The metabolism of CPT-11 by CYP3A5 was markedly different because neither APC or NPC nor M2 was produced, whereas only one new metabolite, M4 (molecular weight, 558), was generated by de-ethylation of the CPT moiety. Irinotecan 18-24 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 28-34 10815927-11 2000 The parameters of CPT-11 biotransformation into M2 and M4 were examined using cell lines expressing, respectively, with CYP3A4 and CYP3A5, indicating that CPT-11 is preferentially metabolized by CYP3A4. Irinotecan 18-24 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 120-126 10815927-11 2000 The parameters of CPT-11 biotransformation into M2 and M4 were examined using cell lines expressing, respectively, with CYP3A4 and CYP3A5, indicating that CPT-11 is preferentially metabolized by CYP3A4. Irinotecan 18-24 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 131-137 10815927-11 2000 The parameters of CPT-11 biotransformation into M2 and M4 were examined using cell lines expressing, respectively, with CYP3A4 and CYP3A5, indicating that CPT-11 is preferentially metabolized by CYP3A4. Irinotecan 18-24 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 195-201 10815927-11 2000 The parameters of CPT-11 biotransformation into M2 and M4 were examined using cell lines expressing, respectively, with CYP3A4 and CYP3A5, indicating that CPT-11 is preferentially metabolized by CYP3A4. Irinotecan 155-161 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 131-137 10815927-11 2000 The parameters of CPT-11 biotransformation into M2 and M4 were examined using cell lines expressing, respectively, with CYP3A4 and CYP3A5, indicating that CPT-11 is preferentially metabolized by CYP3A4. Irinotecan 155-161 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 195-201 10815927-12 2000 In conclusion, CYP3A plays a major role in the metabolism of CPT-11, with some differences of the metabolic profile exhibited by 3A4 and 3A5. Irinotecan 61-67 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 15-20 10838661-10 2000 Collectively, the results suggest that augmentation of the therapeutic efficacy of irinotecan against colon cancer liver metastases by immunomodulation with JBT 3002 may be associated with elevated inducible nitric oxide synthase and endogenous interleukin-15 in tumor-infiltrating macrophages. Irinotecan 83-93 nitric oxide synthase 2, inducible Mus musculus 198-229 10838661-10 2000 Collectively, the results suggest that augmentation of the therapeutic efficacy of irinotecan against colon cancer liver metastases by immunomodulation with JBT 3002 may be associated with elevated inducible nitric oxide synthase and endogenous interleukin-15 in tumor-infiltrating macrophages. Irinotecan 83-93 interleukin 15 Mus musculus 245-259 10933967-6 2000 Specifically, IMR32.ODC.CE cells expressed approximately eightfold more CE activity than IMR32.CMV.neo cells; and 5 microM CPT-11 reduced the clonogenic potential of IMR32.ODC.CE cells to zero, while 50 microM CPT-11 was required to produce the same effect with IMR32.CMV.neo cells. Irinotecan 123-129 ornithine decarboxylase 1 Homo sapiens 20-23 10933967-6 2000 Specifically, IMR32.ODC.CE cells expressed approximately eightfold more CE activity than IMR32.CMV.neo cells; and 5 microM CPT-11 reduced the clonogenic potential of IMR32.ODC.CE cells to zero, while 50 microM CPT-11 was required to produce the same effect with IMR32.CMV.neo cells. Irinotecan 123-129 ornithine decarboxylase 1 Homo sapiens 172-175 10738246-0 2000 Ectopic p16(ink4) expression enhances CPT-11-induced apoptosis through increased delay in S-phase progression in human non-small-cell-lung-cancer cells. Irinotecan 38-44 cyclin dependent kinase inhibitor 2A Homo sapiens 8-11 10738246-0 2000 Ectopic p16(ink4) expression enhances CPT-11-induced apoptosis through increased delay in S-phase progression in human non-small-cell-lung-cancer cells. Irinotecan 38-44 cyclin dependent kinase inhibitor 2A Homo sapiens 12-16 10738246-2 2000 We have found that ectopic p16(INK4) expression increased cellular sensitivity of human non-small-cell-lung-cancer (NSCLC) A549 cells to a selective growth-inhibitory effect induced by the topoisomerase-I inhibitor 11, 7-ethyl-10-[4-(1-piperidino)-1-piperidino] carbonyloxy camptothecin (CPT-11) in vitro. Irinotecan 288-294 cyclin dependent kinase inhibitor 2A Homo sapiens 27-30 10738246-3 2000 In this study, we observed enhanced apoptosis characterized by DNA fragmentation in A549 cells transfected with p16(INK4) cDNA (A549/p16-1) and treated with CPT-11. Irinotecan 157-163 cyclin dependent kinase inhibitor 2A Homo sapiens 112-115 10738246-3 2000 In this study, we observed enhanced apoptosis characterized by DNA fragmentation in A549 cells transfected with p16(INK4) cDNA (A549/p16-1) and treated with CPT-11. Irinotecan 157-163 cyclin dependent kinase inhibitor 2A Homo sapiens 116-120 10738246-3 2000 In this study, we observed enhanced apoptosis characterized by DNA fragmentation in A549 cells transfected with p16(INK4) cDNA (A549/p16-1) and treated with CPT-11. Irinotecan 157-163 cyclin dependent kinase inhibitor 2A Homo sapiens 133-136 10738246-5 2000 In A549/p16-1 cells, cytosolic peptidase activities that cleaved Z-DEVD-7-amino-4-trifluoromethylcoumarin increased during CPT-11-induced apoptosis and were suppressed by a highly specific caspase-3 and caspase-3-like inhibitor, Z-DEVD-fluoromethylketone. Irinotecan 123-129 cyclin dependent kinase inhibitor 2A Homo sapiens 8-11 10738246-5 2000 In A549/p16-1 cells, cytosolic peptidase activities that cleaved Z-DEVD-7-amino-4-trifluoromethylcoumarin increased during CPT-11-induced apoptosis and were suppressed by a highly specific caspase-3 and caspase-3-like inhibitor, Z-DEVD-fluoromethylketone. Irinotecan 123-129 caspase 3 Sus scrofa 189-198 10738246-5 2000 In A549/p16-1 cells, cytosolic peptidase activities that cleaved Z-DEVD-7-amino-4-trifluoromethylcoumarin increased during CPT-11-induced apoptosis and were suppressed by a highly specific caspase-3 and caspase-3-like inhibitor, Z-DEVD-fluoromethylketone. Irinotecan 123-129 caspase 3 Homo sapiens 203-212 10738246-6 2000 These findings indicate that p16(INK) is positively involved in the activation pathway of the caspase-3 induced by CPT-11. Irinotecan 115-121 cyclin dependent kinase inhibitor 2A Homo sapiens 29-32 10738246-6 2000 These findings indicate that p16(INK) is positively involved in the activation pathway of the caspase-3 induced by CPT-11. Irinotecan 115-121 caspase 3 Homo sapiens 94-103 10738246-7 2000 The increased delay in S-phase progression and subsequent induction of apoptosis were observed in CPT-11-treated A549/p16-1 cells on the basis of DNA histograms. Irinotecan 98-104 cyclin dependent kinase inhibitor 2A Homo sapiens 118-121 10738246-8 2000 Specific down-regulation of the cyclin-A protein level in A549/p16-1 cells was observed after CPT-11-treatment, whereas cyclin B, cdk2, and cdc2 protein levels were unaffected. Irinotecan 94-100 proliferating cell nuclear antigen Homo sapiens 32-38 10738246-9 2000 These results suggest that ectopic p16(INK4) expression inappropriately decreases cyclin A and thereby terminates CPT-11-induced G(2)/M accumulation, which is followed by increased apoptosis in p16(INK4)-expressing A549 cells. Irinotecan 114-120 cyclin dependent kinase inhibitor 2A Homo sapiens 35-38 10738246-9 2000 These results suggest that ectopic p16(INK4) expression inappropriately decreases cyclin A and thereby terminates CPT-11-induced G(2)/M accumulation, which is followed by increased apoptosis in p16(INK4)-expressing A549 cells. Irinotecan 114-120 cyclin dependent kinase inhibitor 2A Homo sapiens 39-43 10738246-9 2000 These results suggest that ectopic p16(INK4) expression inappropriately decreases cyclin A and thereby terminates CPT-11-induced G(2)/M accumulation, which is followed by increased apoptosis in p16(INK4)-expressing A549 cells. Irinotecan 114-120 cyclin dependent kinase inhibitor 2A Homo sapiens 198-202 10728672-4 2000 hCE-2 has a 12.5-fold higher affinity for CPT-11 and a 5-fold higher maximal rate of CPT-11 hydrolysis when compared with hCE-1. Irinotecan 42-48 carboxylesterase 2 Homo sapiens 0-5 10728672-4 2000 hCE-2 has a 12.5-fold higher affinity for CPT-11 and a 5-fold higher maximal rate of CPT-11 hydrolysis when compared with hCE-1. Irinotecan 85-91 carboxylesterase 2 Homo sapiens 0-5 10728672-5 2000 In cytotoxicity assays, incubation of 1 microM CPT-11 with hCE-2 (3.6 microg/ml) resulted in a 60% reduction in survival of SQ20b cells. Irinotecan 47-53 carboxylesterase 2 Homo sapiens 59-64 10728672-6 2000 No significant reduction in cell survival was observed after incubation of CPT-11 with hCE-1. Irinotecan 75-81 RNA guanylyltransferase and 5'-phosphatase Homo sapiens 87-92 10728672-8 2000 hCE-2 likely plays a substantial role in CPT-11 activation in human liver at relevant pharmacological concentrations. Irinotecan 41-47 carboxylesterase 2 Homo sapiens 0-5 10690558-5 2000 Compared to A253/Vec, A253/Bax cells exhibited 9.5- and 13.5-fold increases in sensitivity to raltitrexed and SN-38, respectively. Irinotecan 110-115 BCL2 associated X, apoptosis regulator Homo sapiens 27-30 10741701-0 2000 Pharmacokinetics of irinotecan and its metabolites SN-38 and APC in children with recurrent solid tumors after protracted low-dose irinotecan. Irinotecan 20-30 APC regulator of WNT signaling pathway Homo sapiens 61-64 10741701-0 2000 Pharmacokinetics of irinotecan and its metabolites SN-38 and APC in children with recurrent solid tumors after protracted low-dose irinotecan. Irinotecan 131-141 APC regulator of WNT signaling pathway Homo sapiens 61-64 10732766-0 2000 Sensitivity to CPT-11 of xenografted human colorectal cancers as a function of microsatellite instability and p53 status. Irinotecan 15-21 tumor protein p53 Homo sapiens 110-113 10732766-16 2000 Taken together, these observations indicate that MSI and p53 alterations could be associated with different CPT-11 sensitivities; MSI phenotype moderately influences the CPT-11 sensitivity, MSI+ being more sensitive than MSI(-)CRC freshly obtained from patients, mutp53 status being associated with a poor response to CPT-11. Irinotecan 108-114 RB binding protein 4, chromatin remodeling factor Homo sapiens 49-52 10732766-16 2000 Taken together, these observations indicate that MSI and p53 alterations could be associated with different CPT-11 sensitivities; MSI phenotype moderately influences the CPT-11 sensitivity, MSI+ being more sensitive than MSI(-)CRC freshly obtained from patients, mutp53 status being associated with a poor response to CPT-11. Irinotecan 108-114 tumor protein p53 Homo sapiens 57-60 10732766-16 2000 Taken together, these observations indicate that MSI and p53 alterations could be associated with different CPT-11 sensitivities; MSI phenotype moderately influences the CPT-11 sensitivity, MSI+ being more sensitive than MSI(-)CRC freshly obtained from patients, mutp53 status being associated with a poor response to CPT-11. Irinotecan 170-176 RB binding protein 4, chromatin remodeling factor Homo sapiens 130-133 10732766-16 2000 Taken together, these observations indicate that MSI and p53 alterations could be associated with different CPT-11 sensitivities; MSI phenotype moderately influences the CPT-11 sensitivity, MSI+ being more sensitive than MSI(-)CRC freshly obtained from patients, mutp53 status being associated with a poor response to CPT-11. Irinotecan 170-176 RB binding protein 4, chromatin remodeling factor Homo sapiens 130-133 10732766-16 2000 Taken together, these observations indicate that MSI and p53 alterations could be associated with different CPT-11 sensitivities; MSI phenotype moderately influences the CPT-11 sensitivity, MSI+ being more sensitive than MSI(-)CRC freshly obtained from patients, mutp53 status being associated with a poor response to CPT-11. Irinotecan 170-176 RB binding protein 4, chromatin remodeling factor Homo sapiens 130-133 10732775-4 2000 In this report, we demonstrated that apoptosis induced by 7-ethyl-10-hydroxycamptothecin, tumour necrosis factor-alpha (TNF-alpha) or interleukin (IL)-2-activated natural killer (NK) cells was significantly inhibited in tumour cells transduced with the SCC Ag-1 cDNA, as compared to control cells in vitro. Irinotecan 58-88 serpin family B member 3 Homo sapiens 253-256 10770640-11 2000 These findings suggest that CPT-11 and 5-FU may thus be useful as possible anticancer agents for use in a combination therapy regimen using wt p53 gene transfer. Irinotecan 28-34 tumor protein p53 Homo sapiens 143-146 10690558-12 2000 In contrast, both raltitrexed and irinotecan were significantly more active against A253/Bax xenografts than against A253/Vec xenografts; the yield for complete tumor regression (cure) was 40% and 100% with raltitrexed and irinotecan, respectively, with no significant toxicity. Irinotecan 34-44 BCL2 associated X, apoptosis regulator Homo sapiens 89-92 10646841-0 2000 Therapy of human pancreatic carcinoma implants by irinotecan and the oral immunomodulator JBT 3002 is associated with enhanced expression of inducible nitric oxide synthase in tumor-infiltrating macrophages. Irinotecan 50-60 nitric oxide synthase 2 Homo sapiens 141-172 10639573-6 2000 Differing from MMC, MGMT was shown to participate in the resistance of Topo I inhibitors (CPT-11, SN-38 and DX-8951f), while GSTpi and MDR1 were involved in docetaxel (TXT) resistance. Irinotecan 90-96 O-6-methylguanine-DNA methyltransferase Homo sapiens 20-24 10639573-6 2000 Differing from MMC, MGMT was shown to participate in the resistance of Topo I inhibitors (CPT-11, SN-38 and DX-8951f), while GSTpi and MDR1 were involved in docetaxel (TXT) resistance. Irinotecan 98-103 O-6-methylguanine-DNA methyltransferase Homo sapiens 20-24 10646841-11 2000 injection of CPT-11 can decrease the growth of human pancreatic carcinoma and the incidence of metastasis in nude mice by both a direct antitumor effect and the activation of iNOS in infiltrating macrophages. Irinotecan 13-19 nitric oxide synthase 2, inducible Mus musculus 175-179 10854139-5 2000 Gemcitabine and to a lesser extent irinotecan inhibited the secretion of the proinflammatory cytokine IL-6 at concentrations of each drug that produced only small decreases in cell viability. Irinotecan 35-45 interleukin 6 Homo sapiens 102-106 10803918-10 2000 The combined treatment of irradiation and SN-38 showed supraadditive effects in all four cell lines independent of their p53 status. Irinotecan 42-47 transformation related protein 53, pseudogene Mus musculus 121-124 10803925-1 2000 PURPOSE: To investigate the effects of several camptothecin analogs including 9-aminocamptothecin (9-AC), SN38, topotecan, and irinotecan (CPT-11) on the enzymes involved in the pyrimidine salvage pathway including thymidylate synthase (TS). Irinotecan 139-145 thymidylate synthetase Homo sapiens 215-235 10803925-1 2000 PURPOSE: To investigate the effects of several camptothecin analogs including 9-aminocamptothecin (9-AC), SN38, topotecan, and irinotecan (CPT-11) on the enzymes involved in the pyrimidine salvage pathway including thymidylate synthase (TS). Irinotecan 139-145 thymidylate synthetase Homo sapiens 237-239 10854139-7 2000 Higher doses of gemcitabine and irinotecan caused a surge in IL-6 release and this was not due to release of intracellular stores of IL-6 through lysis of the cells. Irinotecan 32-42 interleukin 6 Homo sapiens 61-65 10854139-8 2000 CONCLUSIONS: These results suggest that irinotecan and gemcitabine are not only more likely to be active against mesothelioma than other new chemotherapy agents but may also produce a palliative effect in nonresponders to these agents by decreasing the secretion of IL-6. Irinotecan 40-50 interleukin 6 Homo sapiens 266-270 10626806-0 1999 Tumor necrosis factor-related apoptosis-inducing ligand"s antitumor activity in vivo is enhanced by the chemotherapeutic agent CPT-11. Irinotecan 127-133 TNF superfamily member 10 Homo sapiens 0-55 10626806-6 1999 Moreover, the combination of TRAIL and CPT-11, a water-soluble analogue of camptothecin, greatly enhanced the antitumor activity of TRAIL in vivo. Irinotecan 39-45 TNF superfamily member 10 Homo sapiens 132-137 10626806-7 1999 TRAIL plus CPT-11 treatment of both 3- and 10-day established TRAIL-sensitive tumors resulted in both a significant inhibition of tumor growth and a high proportion of complete tumor regressions. Irinotecan 11-17 TNF superfamily member 10 Homo sapiens 62-67 10626806-8 1999 Treatment of TRAIL-resistant tumors with TRAIL and CPT-11 dramatically slowed tumor growth and induced a transient tumor regression. Irinotecan 51-57 TNF superfamily member 10 Homo sapiens 13-18 10626806-9 1999 These data demonstrate that TRAIL alone is a potent antitumor agent in vivo, and its activity can be significantly enhanced in combination with the chemotherapeutic agent CPT-11. Irinotecan 171-177 TNF superfamily member 10 Homo sapiens 28-33 10570064-11 1999 Also, preliminary molecular modeling of the interaction between AChE and CPT-11 indicated that the latter does not bind at the same site as tacrine. Irinotecan 73-79 acetylcholinesterase (Cartwright blood group) Homo sapiens 64-68 10604724-0 1999 Schedule-dependent cytotoxicity of SN-38 in p53 wild-type and mutant colon adenocarcinoma cell lines. Irinotecan 35-40 tumor protein p53 Homo sapiens 44-47 10604724-10 1999 Dose-dependent p53-associated G1 arrest, in the absence of DNA synthesis indicates an additional cytotoxic mechanism for SN-38, which requires higher concentrations than the S phase mechanism, and detection of which seems to involve p53. Irinotecan 121-126 tumor protein p53 Homo sapiens 15-18 10604724-12 1999 Although expression of wild-type p53 leads to a more rapid induction of apoptosis, SN-38 cytotoxicity was generally greater in cells with mutant p53, as wild-type cells escaped apoptosis by p53 associated prolonged cell cycle arrest. Irinotecan 83-88 tumor protein p53 Homo sapiens 145-148 10604724-12 1999 Although expression of wild-type p53 leads to a more rapid induction of apoptosis, SN-38 cytotoxicity was generally greater in cells with mutant p53, as wild-type cells escaped apoptosis by p53 associated prolonged cell cycle arrest. Irinotecan 83-88 tumor protein p53 Homo sapiens 145-148 10570064-13 1999 The rapid reversibility of the inhibition of AChE by CPT-11 and the lower activity of the carboxylate form are likely reasons for the transient nature of the cholinergic toxicity observed clinically. Irinotecan 53-59 acetylcholinesterase (Cartwright blood group) Homo sapiens 45-49 10411544-10 1999 Apo2L cooperated synergistically with the chemotherapeutic drugs 5-fluorouracil or CPT-11, causing substantial tumor regression or complete tumor ablation. Irinotecan 83-89 tumor necrosis factor (ligand) superfamily, member 10 Mus musculus 0-5 10569939-1 1999 Homocamptothecin (hCPT) contains a seven-membered beta-hydroxylactone in place of the conventional six-membered alpha-hydroxylactone ring found in camptothecin and its tumor active analogues, including topotecan and irinotecan. Irinotecan 216-226 choline phosphotransferase 1 Homo sapiens 18-22 10493507-4 1999 Furthermore, BCRP/MXR/ABCP expression levels in various partially revertant T8 cells correlated with the levels of resistance to topotecan, SN-38, and mitoxantrone, strongly suggesting BCRP/MXR/ABCP to be the transporter responsible for the enhanced efflux. Irinotecan 140-145 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 13-17 10493507-4 1999 Furthermore, BCRP/MXR/ABCP expression levels in various partially revertant T8 cells correlated with the levels of resistance to topotecan, SN-38, and mitoxantrone, strongly suggesting BCRP/MXR/ABCP to be the transporter responsible for the enhanced efflux. Irinotecan 140-145 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 18-21 10493507-4 1999 Furthermore, BCRP/MXR/ABCP expression levels in various partially revertant T8 cells correlated with the levels of resistance to topotecan, SN-38, and mitoxantrone, strongly suggesting BCRP/MXR/ABCP to be the transporter responsible for the enhanced efflux. Irinotecan 140-145 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 22-26 10493507-4 1999 Furthermore, BCRP/MXR/ABCP expression levels in various partially revertant T8 cells correlated with the levels of resistance to topotecan, SN-38, and mitoxantrone, strongly suggesting BCRP/MXR/ABCP to be the transporter responsible for the enhanced efflux. Irinotecan 140-145 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 185-189 10493507-4 1999 Furthermore, BCRP/MXR/ABCP expression levels in various partially revertant T8 cells correlated with the levels of resistance to topotecan, SN-38, and mitoxantrone, strongly suggesting BCRP/MXR/ABCP to be the transporter responsible for the enhanced efflux. Irinotecan 140-145 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 190-193 10493507-4 1999 Furthermore, BCRP/MXR/ABCP expression levels in various partially revertant T8 cells correlated with the levels of resistance to topotecan, SN-38, and mitoxantrone, strongly suggesting BCRP/MXR/ABCP to be the transporter responsible for the enhanced efflux. Irinotecan 140-145 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 194-198 10416605-5 1999 The glycinate esters are also 20-40-fold less potent than CPT-11 in inhibiting human acetylcholinesterase. Irinotecan 58-64 acetylcholinesterase (Cartwright blood group) Homo sapiens 85-105 10606256-2 1999 The most extensively studied of these has been irinotecan (CPT-II). Irinotecan 47-57 carnitine palmitoyltransferase 2 Homo sapiens 59-65 10695015-5 1999 Human cMOAT cDNA transfected LLC-PK1 cells, LLC/cMOAT-1, have increased resistance to vincristine (VCR), 7-ethyl-10-hydroxycamptothecin (SN-38), and cisplatin. Irinotecan 105-135 ATP binding cassette subfamily C member 2 Homo sapiens 6-11 10695015-5 1999 Human cMOAT cDNA transfected LLC-PK1 cells, LLC/cMOAT-1, have increased resistance to vincristine (VCR), 7-ethyl-10-hydroxycamptothecin (SN-38), and cisplatin. Irinotecan 137-142 ATP binding cassette subfamily C member 2 Homo sapiens 6-11 10695015-6 1999 The multidrug resistance (MDR)-reversing agents, cyclosporin A (CsA) and PAK-104P, almost completely reversed the resistance to VCR, SN-38 and cisplatin of LLC/cMOAT-1 cells by interacting with the substrate binding site of cMOAT. Irinotecan 133-138 ATP-binding cassette, sub-family C (CFTR/MRP), member 2 Mus musculus 160-165 10473099-0 1999 Oral administration of the immunomodulator JBT-3002 induces endogenous interleukin 15 in intestinal macrophages for protection against irinotecan-mediated destruction of intestinal epithelium. Irinotecan 135-145 interleukin 15 Mus musculus 71-85 10473099-10 1999 Immunohistochemical analyses of the intestines of mice treated with JBT-3002 and CPT-11 demonstrated an increase in the number of dividing cells in the crypts and enhanced expression of IL-15 in lamina propria cells; the increase correlated with increased expression of the IL-15 gene as determined by semiquantitative reverse transcriptase-PCR. Irinotecan 81-87 interleukin 15 Mus musculus 186-191 10473099-10 1999 Immunohistochemical analyses of the intestines of mice treated with JBT-3002 and CPT-11 demonstrated an increase in the number of dividing cells in the crypts and enhanced expression of IL-15 in lamina propria cells; the increase correlated with increased expression of the IL-15 gene as determined by semiquantitative reverse transcriptase-PCR. Irinotecan 81-87 interleukin 15 Mus musculus 274-279 10473099-12 1999 Moreover, the presence of IL-15 decreased irinotecan-mediated cytotoxicity of IEC-6 epithelial cells. Irinotecan 42-52 interleukin 15 Rattus norvegicus 26-31 10473099-14 1999 administration of JBT-3002 induces expression of IL-15 by macrophages in the lamina propria, which can prevent irinotecan-induced injury to the intestinal mucosa. Irinotecan 111-121 interleukin 15 Mus musculus 49-54 10381366-0 1999 Glucuronidation of 7-ethyl-10-hydroxycamptothecin (SN-38) by the human UDP-glucuronosyltransferases encoded at the UGT1 locus. Irinotecan 19-49 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 115-119 10381366-0 1999 Glucuronidation of 7-ethyl-10-hydroxycamptothecin (SN-38) by the human UDP-glucuronosyltransferases encoded at the UGT1 locus. Irinotecan 51-56 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 115-119 10381366-3 1999 In this study, we examined the potential for each of the UGT1-encoded isoforms (UGT1A1 and UGT1A3 through UGT1A10) to glucuronidate SN-38. Irinotecan 132-137 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 57-61 10381366-3 1999 In this study, we examined the potential for each of the UGT1-encoded isoforms (UGT1A1 and UGT1A3 through UGT1A10) to glucuronidate SN-38. Irinotecan 132-137 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 80-86 10381366-3 1999 In this study, we examined the potential for each of the UGT1-encoded isoforms (UGT1A1 and UGT1A3 through UGT1A10) to glucuronidate SN-38. Irinotecan 132-137 UDP glucuronosyltransferase family 1 member A3 Homo sapiens 91-97 10381366-3 1999 In this study, we examined the potential for each of the UGT1-encoded isoforms (UGT1A1 and UGT1A3 through UGT1A10) to glucuronidate SN-38. Irinotecan 132-137 UDP glucuronosyltransferase family 1 member A10 Homo sapiens 106-113 10381366-8 1999 UGT1A7 glucuronidates SN-38 with an apparent Km of 5 microM. Irinotecan 22-27 UDP glucuronosyltransferase family 1 member A7 Homo sapiens 0-6 10340924-0 1999 Phenotype-genotype correlation of in vitro SN-38 (active metabolite of irinotecan) and bilirubin glucuronidation in human liver tissue with UGT1A1 promoter polymorphism. Irinotecan 43-48 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 140-146 10340924-0 1999 Phenotype-genotype correlation of in vitro SN-38 (active metabolite of irinotecan) and bilirubin glucuronidation in human liver tissue with UGT1A1 promoter polymorphism. Irinotecan 71-81 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 140-146 10340924-1 1999 BACKGROUND: Hepatic uridine diphosphate glucuronosyltransferase (UGT) isoform 1A1 (UGT1A1) is primarily responsible for the glucuronidation of SN-38 (7-ethyl-10-hydroxycamptothecin), the active metabolite of the anticancer agent irinotecan. Irinotecan 143-148 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 83-89 10340924-1 1999 BACKGROUND: Hepatic uridine diphosphate glucuronosyltransferase (UGT) isoform 1A1 (UGT1A1) is primarily responsible for the glucuronidation of SN-38 (7-ethyl-10-hydroxycamptothecin), the active metabolite of the anticancer agent irinotecan. Irinotecan 150-180 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 83-89 10340924-1 1999 BACKGROUND: Hepatic uridine diphosphate glucuronosyltransferase (UGT) isoform 1A1 (UGT1A1) is primarily responsible for the glucuronidation of SN-38 (7-ethyl-10-hydroxycamptothecin), the active metabolite of the anticancer agent irinotecan. Irinotecan 229-239 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 83-89 10220571-0 1999 ATP-Dependent efflux of CPT-11 and SN-38 by the multidrug resistance protein (MRP) and its inhibition by PAK-104P. Irinotecan 24-30 ATP binding cassette subfamily C member 1 Homo sapiens 48-76 10410150-1 1999 Two phase I studies (CIC-therapy) were conducted in advanced non-small cell lung cancer (NSCLC) to determine the maximum tolerable dose (MTD) of CPT-11 combined with cisplatin and ifosfamide, and MTD of cisplatin combined with CPT-11 and ifosfamide with G-CSF support, respectively. Irinotecan 145-151 colony stimulating factor 3 Homo sapiens 254-259 10220571-0 1999 ATP-Dependent efflux of CPT-11 and SN-38 by the multidrug resistance protein (MRP) and its inhibition by PAK-104P. Irinotecan 24-30 ATP binding cassette subfamily C member 1 Homo sapiens 78-81 10220571-0 1999 ATP-Dependent efflux of CPT-11 and SN-38 by the multidrug resistance protein (MRP) and its inhibition by PAK-104P. Irinotecan 35-40 ATP binding cassette subfamily C member 1 Homo sapiens 48-76 10220571-0 1999 ATP-Dependent efflux of CPT-11 and SN-38 by the multidrug resistance protein (MRP) and its inhibition by PAK-104P. Irinotecan 35-40 ATP binding cassette subfamily C member 1 Homo sapiens 78-81 10220571-3 1999 To see whether MRP is involved in CPT-11 and SN-38 resistance, MRP cDNA was transfected into KB-3-1 cells. Irinotecan 45-50 ATP binding cassette subfamily C member 1 Homo sapiens 15-18 10220571-4 1999 The transfectant, KB/MRP, which overexpressed MRP, was resistant to both CPT-11 and SN-38. Irinotecan 73-79 ATP binding cassette subfamily C member 1 Homo sapiens 21-24 10220571-4 1999 The transfectant, KB/MRP, which overexpressed MRP, was resistant to both CPT-11 and SN-38. Irinotecan 73-79 ATP binding cassette subfamily C member 1 Homo sapiens 46-49 10220571-4 1999 The transfectant, KB/MRP, which overexpressed MRP, was resistant to both CPT-11 and SN-38. Irinotecan 84-89 ATP binding cassette subfamily C member 1 Homo sapiens 21-24 10220571-4 1999 The transfectant, KB/MRP, which overexpressed MRP, was resistant to both CPT-11 and SN-38. Irinotecan 84-89 ATP binding cassette subfamily C member 1 Homo sapiens 46-49 10220571-6 1999 The accumulation of both CPT-11 and SN-38 in C-A120 and KB/MRP cells was lower than that in KB-3-1 cells. Irinotecan 25-31 ATP binding cassette subfamily C member 1 Homo sapiens 59-62 10220571-6 1999 The accumulation of both CPT-11 and SN-38 in C-A120 and KB/MRP cells was lower than that in KB-3-1 cells. Irinotecan 36-41 ATP binding cassette subfamily C member 1 Homo sapiens 59-62 10220571-7 1999 The treatment with 10 microM PAK-104P increased the accumulation of CPT-11 and SN-38 in C-A120 and KB/MRP cells to a level similar to that found in KB-3-1 cells. Irinotecan 68-74 ATP binding cassette subfamily C member 1 Homo sapiens 102-105 10220571-7 1999 The treatment with 10 microM PAK-104P increased the accumulation of CPT-11 and SN-38 in C-A120 and KB/MRP cells to a level similar to that found in KB-3-1 cells. Irinotecan 79-84 ATP binding cassette subfamily C member 1 Homo sapiens 102-105 10220571-8 1999 The ATP-dependent efflux of CPT-11 and SN-38 from C-A120 and KB/MRP cells was inhibited by PAK-104P. Irinotecan 28-34 ATP binding cassette subfamily C member 1 Homo sapiens 64-67 10220571-8 1999 The ATP-dependent efflux of CPT-11 and SN-38 from C-A120 and KB/MRP cells was inhibited by PAK-104P. Irinotecan 39-44 ATP binding cassette subfamily C member 1 Homo sapiens 64-67 10220571-11 1999 These findings suggest that MRP transports CPT-11 and SN-38 and is involved in resistance to CPT-11 and SN-38 and that PAK-104P reverses the resistance to CPT-11 and SN-38 in tumors that overexpress MRP. Irinotecan 43-49 ATP binding cassette subfamily C member 1 Homo sapiens 28-31 10220571-11 1999 These findings suggest that MRP transports CPT-11 and SN-38 and is involved in resistance to CPT-11 and SN-38 and that PAK-104P reverses the resistance to CPT-11 and SN-38 in tumors that overexpress MRP. Irinotecan 43-49 ATP binding cassette subfamily C member 1 Homo sapiens 199-202 10220571-11 1999 These findings suggest that MRP transports CPT-11 and SN-38 and is involved in resistance to CPT-11 and SN-38 and that PAK-104P reverses the resistance to CPT-11 and SN-38 in tumors that overexpress MRP. Irinotecan 54-59 ATP binding cassette subfamily C member 1 Homo sapiens 28-31 10220571-11 1999 These findings suggest that MRP transports CPT-11 and SN-38 and is involved in resistance to CPT-11 and SN-38 and that PAK-104P reverses the resistance to CPT-11 and SN-38 in tumors that overexpress MRP. Irinotecan 54-59 ATP binding cassette subfamily C member 1 Homo sapiens 199-202 10220571-11 1999 These findings suggest that MRP transports CPT-11 and SN-38 and is involved in resistance to CPT-11 and SN-38 and that PAK-104P reverses the resistance to CPT-11 and SN-38 in tumors that overexpress MRP. Irinotecan 93-99 ATP binding cassette subfamily C member 1 Homo sapiens 28-31 10220571-11 1999 These findings suggest that MRP transports CPT-11 and SN-38 and is involved in resistance to CPT-11 and SN-38 and that PAK-104P reverses the resistance to CPT-11 and SN-38 in tumors that overexpress MRP. Irinotecan 104-109 ATP binding cassette subfamily C member 1 Homo sapiens 28-31 10220571-11 1999 These findings suggest that MRP transports CPT-11 and SN-38 and is involved in resistance to CPT-11 and SN-38 and that PAK-104P reverses the resistance to CPT-11 and SN-38 in tumors that overexpress MRP. Irinotecan 93-99 ATP binding cassette subfamily C member 1 Homo sapiens 28-31 10220571-11 1999 These findings suggest that MRP transports CPT-11 and SN-38 and is involved in resistance to CPT-11 and SN-38 and that PAK-104P reverses the resistance to CPT-11 and SN-38 in tumors that overexpress MRP. Irinotecan 104-109 ATP binding cassette subfamily C member 1 Homo sapiens 28-31 10197614-0 1999 The anticancer prodrug CPT-11 is a potent inhibitor of acetylcholinesterase but is rapidly catalyzed to SN-38 by butyrylcholinesterase. Irinotecan 23-29 acetylcholinesterase (Cartwright blood group) Homo sapiens 55-75 10378768-13 1999 These findings suggested that an unknown transporter distinct from P-gp, MRP or cMOAT is expressed in KCP-4 cells and transports CPT-11 and SN-38. Irinotecan 129-135 ATP binding cassette subfamily B member 1 Homo sapiens 67-71 10378768-13 1999 These findings suggested that an unknown transporter distinct from P-gp, MRP or cMOAT is expressed in KCP-4 cells and transports CPT-11 and SN-38. Irinotecan 129-135 ATP binding cassette subfamily C member 1 Homo sapiens 73-76 10378768-13 1999 These findings suggested that an unknown transporter distinct from P-gp, MRP or cMOAT is expressed in KCP-4 cells and transports CPT-11 and SN-38. Irinotecan 129-135 ATP binding cassette subfamily C member 2 Homo sapiens 80-85 10378768-13 1999 These findings suggested that an unknown transporter distinct from P-gp, MRP or cMOAT is expressed in KCP-4 cells and transports CPT-11 and SN-38. Irinotecan 140-145 ATP binding cassette subfamily B member 1 Homo sapiens 67-71 10378768-13 1999 These findings suggested that an unknown transporter distinct from P-gp, MRP or cMOAT is expressed in KCP-4 cells and transports CPT-11 and SN-38. Irinotecan 140-145 ATP binding cassette subfamily C member 1 Homo sapiens 73-76 10378768-13 1999 These findings suggested that an unknown transporter distinct from P-gp, MRP or cMOAT is expressed in KCP-4 cells and transports CPT-11 and SN-38. Irinotecan 140-145 ATP binding cassette subfamily C member 2 Homo sapiens 80-85 10197614-2 1999 Although CPT-11 was not a substrate for acetylcholinesterase (AcChE), in vitro assays confirmed that CPT-11 inhibited both human and electric eel AcChE with apparent K(i)s of 415 and 194 nM, respectively. Irinotecan 101-107 acetylcholinesterase (Cartwright blood group) Homo sapiens 146-151 10197614-3 1999 In contrast, human or equine butyryl-cholinesterase (BuChE) converted CPT-11 to SN-38 with K(m)s of 42.4 and 44.2 microM for the human and horse BuChE, respectively. Irinotecan 70-76 butyrylcholinesterase Equus caballus 29-51 10197614-3 1999 In contrast, human or equine butyryl-cholinesterase (BuChE) converted CPT-11 to SN-38 with K(m)s of 42.4 and 44.2 microM for the human and horse BuChE, respectively. Irinotecan 70-76 butyrylcholinesterase Equus caballus 53-58 10197614-3 1999 In contrast, human or equine butyryl-cholinesterase (BuChE) converted CPT-11 to SN-38 with K(m)s of 42.4 and 44.2 microM for the human and horse BuChE, respectively. Irinotecan 70-76 butyrylcholinesterase Equus caballus 145-150 10197614-3 1999 In contrast, human or equine butyryl-cholinesterase (BuChE) converted CPT-11 to SN-38 with K(m)s of 42.4 and 44.2 microM for the human and horse BuChE, respectively. Irinotecan 80-85 butyrylcholinesterase Equus caballus 29-51 10197614-3 1999 In contrast, human or equine butyryl-cholinesterase (BuChE) converted CPT-11 to SN-38 with K(m)s of 42.4 and 44.2 microM for the human and horse BuChE, respectively. Irinotecan 80-85 butyrylcholinesterase Equus caballus 53-58 10101137-0 1999 Possible involvement of P-glycoprotein in biliary excretion of CPT-11 in rats. Irinotecan 63-69 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 24-38 10197614-3 1999 In contrast, human or equine butyryl-cholinesterase (BuChE) converted CPT-11 to SN-38 with K(m)s of 42.4 and 44.2 microM for the human and horse BuChE, respectively. Irinotecan 80-85 butyrylcholinesterase Equus caballus 145-150 10101137-1 1999 In our previous work, we found that the biliary excretion of the carboxylate form of irinotecan, CPT-11, on rat bile canalicular membrane consists of two components, the low-affinity one being canalicular multispecific organic anion transporter (cMOAT). Irinotecan 85-95 ATP binding cassette subfamily C member 2 Rattus norvegicus 193-244 10101137-1 1999 In our previous work, we found that the biliary excretion of the carboxylate form of irinotecan, CPT-11, on rat bile canalicular membrane consists of two components, the low-affinity one being canalicular multispecific organic anion transporter (cMOAT). Irinotecan 85-95 ATP binding cassette subfamily C member 2 Rattus norvegicus 246-251 10101137-1 1999 In our previous work, we found that the biliary excretion of the carboxylate form of irinotecan, CPT-11, on rat bile canalicular membrane consists of two components, the low-affinity one being canalicular multispecific organic anion transporter (cMOAT). Irinotecan 97-103 ATP binding cassette subfamily C member 2 Rattus norvegicus 193-244 10197614-4 1999 Modeling of CPT-11 within the predicted active site of AcChE and BuChE corroborated experimental results indicating that, although the drug was oriented correctly for activation, the constraints dictated by the active site gorge were such that CPT-11 would be unlikely to be activated by AcChE. Irinotecan 12-18 acetylcholinesterase (Cartwright blood group) Homo sapiens 55-60 10101137-1 1999 In our previous work, we found that the biliary excretion of the carboxylate form of irinotecan, CPT-11, on rat bile canalicular membrane consists of two components, the low-affinity one being canalicular multispecific organic anion transporter (cMOAT). Irinotecan 97-103 ATP binding cassette subfamily C member 2 Rattus norvegicus 246-251 10197614-4 1999 Modeling of CPT-11 within the predicted active site of AcChE and BuChE corroborated experimental results indicating that, although the drug was oriented correctly for activation, the constraints dictated by the active site gorge were such that CPT-11 would be unlikely to be activated by AcChE. Irinotecan 12-18 acetylcholinesterase (Cartwright blood group) Homo sapiens 288-293 10101137-3 1999 The ATP-dependent uptake of the carboxylate form of CPT-11 was inhibited significantly by several substrates and/or modulators of P-glycoprotein, including PSC-833, verapamil, and cyclosporin A, at a substrate concentration of 5 microM, at which the high-affinity component is involved predominantly in CPT-11 transport. Irinotecan 52-58 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 130-144 10101137-3 1999 The ATP-dependent uptake of the carboxylate form of CPT-11 was inhibited significantly by several substrates and/or modulators of P-glycoprotein, including PSC-833, verapamil, and cyclosporin A, at a substrate concentration of 5 microM, at which the high-affinity component is involved predominantly in CPT-11 transport. Irinotecan 303-309 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 130-144 10197614-4 1999 Modeling of CPT-11 within the predicted active site of AcChE and BuChE corroborated experimental results indicating that, although the drug was oriented correctly for activation, the constraints dictated by the active site gorge were such that CPT-11 would be unlikely to be activated by AcChE. Irinotecan 244-250 acetylcholinesterase (Cartwright blood group) Homo sapiens 55-60 10101137-4 1999 When the concentration of the carboxylate form of CPT-11 was 250 microM, at which the low-affinity component (cMOAT) is involved predominantly in its transport, the inhibitory effect of the above compounds was reduced greatly. Irinotecan 50-56 ATP binding cassette subfamily C member 2 Rattus norvegicus 110-115 10101137-7 1999 These results suggest that P-glycoprotein may act as the high-affinity component in the biliary excretion of the carboxylate form of CPT-11 in rats. Irinotecan 133-139 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 27-41 10197614-4 1999 Modeling of CPT-11 within the predicted active site of AcChE and BuChE corroborated experimental results indicating that, although the drug was oriented correctly for activation, the constraints dictated by the active site gorge were such that CPT-11 would be unlikely to be activated by AcChE. Irinotecan 244-250 butyrylcholinesterase Equus caballus 65-70 10197614-4 1999 Modeling of CPT-11 within the predicted active site of AcChE and BuChE corroborated experimental results indicating that, although the drug was oriented correctly for activation, the constraints dictated by the active site gorge were such that CPT-11 would be unlikely to be activated by AcChE. Irinotecan 244-250 acetylcholinesterase (Cartwright blood group) Homo sapiens 288-293 10100720-5 1999 Results from experiments performed on colon cancer xenografts also show an association between PARP cleavage and response to treatment with TPT or CPT-11. Irinotecan 147-153 poly(ADP-ribose) polymerase 1 Homo sapiens 95-99 10202930-5 1999 We show here that inhibition of NF-kappaB through the adenoviral delivery of a modified form of IkappaBalpha, the inhibitor of NF-kappaB, sensitizes chemoresistant tumors to the apoptotic potential of TNFalpha and of the chemotherapeutic compound CPT-11, resulting in tumor regression. Irinotecan 247-253 nuclear factor kappa B subunit 1 Homo sapiens 32-41 10202930-5 1999 We show here that inhibition of NF-kappaB through the adenoviral delivery of a modified form of IkappaBalpha, the inhibitor of NF-kappaB, sensitizes chemoresistant tumors to the apoptotic potential of TNFalpha and of the chemotherapeutic compound CPT-11, resulting in tumor regression. Irinotecan 247-253 NFKB inhibitor alpha Homo sapiens 96-108 10202930-5 1999 We show here that inhibition of NF-kappaB through the adenoviral delivery of a modified form of IkappaBalpha, the inhibitor of NF-kappaB, sensitizes chemoresistant tumors to the apoptotic potential of TNFalpha and of the chemotherapeutic compound CPT-11, resulting in tumor regression. Irinotecan 247-253 nuclear factor kappa B subunit 1 Homo sapiens 127-136 10202930-5 1999 We show here that inhibition of NF-kappaB through the adenoviral delivery of a modified form of IkappaBalpha, the inhibitor of NF-kappaB, sensitizes chemoresistant tumors to the apoptotic potential of TNFalpha and of the chemotherapeutic compound CPT-11, resulting in tumor regression. Irinotecan 247-253 tumor necrosis factor Homo sapiens 201-209 9918583-7 1999 These results suggest that MRP and P-gp are involved in the active efflux of SN-38 and CPT-11, respectively, from human KB-derived cells. Irinotecan 77-82 ATP binding cassette subfamily C member 3 Homo sapiens 27-30 9918583-7 1999 These results suggest that MRP and P-gp are involved in the active efflux of SN-38 and CPT-11, respectively, from human KB-derived cells. Irinotecan 77-82 ATP binding cassette subfamily B member 1 Homo sapiens 35-39 9918583-7 1999 These results suggest that MRP and P-gp are involved in the active efflux of SN-38 and CPT-11, respectively, from human KB-derived cells. Irinotecan 87-93 ATP binding cassette subfamily C member 3 Homo sapiens 27-30 9918583-7 1999 These results suggest that MRP and P-gp are involved in the active efflux of SN-38 and CPT-11, respectively, from human KB-derived cells. Irinotecan 87-93 ATP binding cassette subfamily B member 1 Homo sapiens 35-39 10500811-6 1999 Moreover CPT and SN-38 induced a strong decrease of mdr1 mRNA in MDR treated cells. Irinotecan 17-22 ATP binding cassette subfamily B member 1 Homo sapiens 52-56 10321506-0 1999 A phase II study of irinotecan and infusional cisplatin with recombinant human granulocyte colony-stimulating factor support for advanced non-small-cell lung cancer. Irinotecan 20-30 colony stimulating factor 3 Homo sapiens 79-116 9786315-1 1998 In six published phase II trials, irinotecan (CPT-II; Camptosar; Pharmacia & Upjohn Co, Kalamazoo, MI) has demonstrated consistent activity with response rates of approximately 13% to 27% in patients with advanced colorectal cancer (CRC) refractory to 5-fluorouracil (5-FU) therapy. Irinotecan 34-44 carnitine palmitoyltransferase 2 Homo sapiens 46-52 9865925-0 1998 Conversion of the CPT-11 metabolite APC to SN-38 by rabbit liver carboxylesterase. Irinotecan 18-24 liver carboxylesterase 1 Oryctolagus cuniculus 59-81 9865925-0 1998 Conversion of the CPT-11 metabolite APC to SN-38 by rabbit liver carboxylesterase. Irinotecan 43-48 liver carboxylesterase 1 Oryctolagus cuniculus 59-81 9865925-2 1998 We report here the conversion of APC (7-ethyl-[4-N-(5-aminopentanoic acid)-1-piperidino] carbonyloxycamptothecin), an inactive metabolite of CPT-11, to SN-38 (7-ethyl-10-hydroxycamptothecin), the active metabolite of CPT-11, by a rabbit liver carboxylesterase. Irinotecan 152-157 liver carboxylesterase 1 Oryctolagus cuniculus 237-259 9766655-5 1998 These E2F-1 transfectants, although resistant to fluoropyrimidines and serum deprivation, were more sensitive to etoposide, doxorubicin, and SN38 (the active metabolite of irinotecan) but not to Taxol. Irinotecan 141-145 E2F transcription factor 1 Homo sapiens 6-11 9766655-5 1998 These E2F-1 transfectants, although resistant to fluoropyrimidines and serum deprivation, were more sensitive to etoposide, doxorubicin, and SN38 (the active metabolite of irinotecan) but not to Taxol. Irinotecan 172-182 E2F transcription factor 1 Homo sapiens 6-11 10489165-3 1999 A253/Bax cells exhibited a significant increase in in vitro sensitivity to various anticancer drugs, including tomudex (9.5-fold), SN-38 (13.8-fold), doxorubicin (7.9-fold), taxol (3.1-fold), 5-FU (2.7-fold), and 5-FU/LV (4.5-fold). Irinotecan 131-136 BCL2 associated X, apoptosis regulator Homo sapiens 5-8 9781601-6 1998 Characteristics of patient subgroups were not different, however, the median survival of patients with elevated c-fos expression from the time of treatment with CPT-11 was 436 days, whereas patients with no detectable c-fos expression had a median survival of 365 days (p = 0.045). Irinotecan 161-167 Fos proto-oncogene, AP-1 transcription factor subunit Homo sapiens 112-117 9786315-1 1998 In six published phase II trials, irinotecan (CPT-II; Camptosar; Pharmacia & Upjohn Co, Kalamazoo, MI) has demonstrated consistent activity with response rates of approximately 13% to 27% in patients with advanced colorectal cancer (CRC) refractory to 5-fluorouracil (5-FU) therapy. Irinotecan 54-63 carnitine palmitoyltransferase 2 Homo sapiens 46-52 9615866-12 1998 Irinotecan undergoes a complex in vivo biotransformation involving several enzyme systems, such as carboxylesterase, UDPGT and cytochrome P450, in children as well as in adults. Irinotecan 0-10 UDP glucuronosyltransferase family 1 member A4 Homo sapiens 117-122 9699654-0 1998 Interleukin 15 offers selective protection from irinotecan-induced intestinal toxicity in a preclinical animal model. Irinotecan 48-58 interleukin 15 Rattus norvegicus 0-14 9699654-3 1998 A rat model with dose-limiting toxicity profiles that are similar to those observed in patients treated with CPT-11 was developed and used to evaluate the role of interleukin 15 (IL-15) in the modulation of the therapeutic selectivity of CPT-11 in normal rats and rats bearing advanced colorectal cancer. Irinotecan 238-244 interleukin 15 Homo sapiens 163-177 9699654-3 1998 A rat model with dose-limiting toxicity profiles that are similar to those observed in patients treated with CPT-11 was developed and used to evaluate the role of interleukin 15 (IL-15) in the modulation of the therapeutic selectivity of CPT-11 in normal rats and rats bearing advanced colorectal cancer. Irinotecan 238-244 interleukin 15 Homo sapiens 179-184 9699654-9 1998 IL-15 offered complete and sustained selective protection against CPT-11-induced delayed diarrhea and lethality. Irinotecan 66-72 interleukin 15 Rattus norvegicus 0-5 9699654-13 1998 The results clearly demonstrated the ability of IL-15 to provide significant protection from CPT-11-induced intestinal toxicity with maintenance of antitumor activity, resulting in an increase in the therapeutic index of CPT-11. Irinotecan 93-99 interleukin 15 Rattus norvegicus 48-53 9699654-13 1998 The results clearly demonstrated the ability of IL-15 to provide significant protection from CPT-11-induced intestinal toxicity with maintenance of antitumor activity, resulting in an increase in the therapeutic index of CPT-11. Irinotecan 221-227 interleukin 15 Rattus norvegicus 48-53 9726087-7 1998 In this model, CPT-11 at a dose 50 mg/kg, daily x5 (MTD) achieved 100% complete tumor regression. Irinotecan 15-21 metallothionein 1E Homo sapiens 52-55 9726089-9 1998 SN-38 is eliminated mainly through conjugation by hepatic uridine glucuronosyltransferase (UGT*1.1), the same isoezyme responsible for glucuronidation of bilirubin. Irinotecan 0-5 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 91-98 9726089-10 1998 Grade 4 irinotecan-related toxicity (ie, neutropenia, diarrhea) has recently been reported in two patients with deficient UGT*1.1 activity. Irinotecan 8-18 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 122-129 9726089-12 1998 Deconjugation of SN-38G to SN-38 by beta-glucuronidase produced by the intestinal flora may contribute to enterohepatic recirculation of SN-38 and delayed intestinal toxicity. Irinotecan 17-22 glucuronidase beta Homo sapiens 36-54 9726089-12 1998 Deconjugation of SN-38G to SN-38 by beta-glucuronidase produced by the intestinal flora may contribute to enterohepatic recirculation of SN-38 and delayed intestinal toxicity. Irinotecan 27-32 glucuronidase beta Homo sapiens 36-54 9726089-12 1998 Deconjugation of SN-38G to SN-38 by beta-glucuronidase produced by the intestinal flora may contribute to enterohepatic recirculation of SN-38 and delayed intestinal toxicity. Irinotecan 27-32 glucuronidase beta Homo sapiens 36-54 9726096-3 1998 A cholinergic syndrome resulting from the inhibition of acetylcholinesterase activity by irinotecan is frequently seen within the first 24 hours after irinotecan administration but is easily controlled with atropine. Irinotecan 89-99 acetylcholinesterase (Cartwright blood group) Homo sapiens 56-76 9726096-3 1998 A cholinergic syndrome resulting from the inhibition of acetylcholinesterase activity by irinotecan is frequently seen within the first 24 hours after irinotecan administration but is easily controlled with atropine. Irinotecan 151-161 acetylcholinesterase (Cartwright blood group) Homo sapiens 56-76 9635592-0 1998 Isolation and partial characterization of a cDNA encoding a rabbit liver carboxylesterase that activates the prodrug irinotecan (CPT-11). Irinotecan 117-127 liver carboxylesterase 1 Oryctolagus cuniculus 67-89 9635592-0 1998 Isolation and partial characterization of a cDNA encoding a rabbit liver carboxylesterase that activates the prodrug irinotecan (CPT-11). Irinotecan 129-135 liver carboxylesterase 1 Oryctolagus cuniculus 67-89 9619871-6 1998 These results suggest that upregulation of bcl-2 in 4-pp-R cells is related to the resistance to CPT-11 as well as to A23187 or dexamethasone. Irinotecan 97-103 BCL2 apoptosis regulator Homo sapiens 43-48 9506540-3 1998 When p53 was expressed at sublethal levels, it sensitized cells to the DNA-damaging drugs Adriamycin, mitomycin C, actinomycin D, etoposide (VP16), cisplatin and CPT11. Irinotecan 162-167 tumor protein p53 Homo sapiens 5-8 9515577-8 1998 When cells were first incubated with CPT-11, the decrease in thymidylate synthase (TS) activity was identical to that obtained after 5FU exposure (1.09 to 0.023 pmol/min/mg protein), but this decrease persisted for 24 hr (0.014 pmol/min/mg protein) (p = 0.035). Irinotecan 37-43 thymidylate synthetase Homo sapiens 61-81 9743293-8 1998 Thus, although p53-dependent apoptosis is induced by camptothecin, topotecan and SN-38 in this human ovarian carcinoma cell line, these drugs induce p53-independent death, as measured by clonogenic assay. Irinotecan 81-86 tumor protein p53 Homo sapiens 15-18 9748119-1 1998 The induction of severe diarrhea limits the usefulness of the DNA topoisomerase I inhibitor irinotecan (CPT-11) in the treatment of advanced colon cancer. Irinotecan 92-102 topoisomerase (DNA) I Mus musculus 62-81 9748119-1 1998 The induction of severe diarrhea limits the usefulness of the DNA topoisomerase I inhibitor irinotecan (CPT-11) in the treatment of advanced colon cancer. Irinotecan 104-110 topoisomerase (DNA) I Mus musculus 62-81 9789606-0 1998 UGT1A1 genotypes and glucuronidation of SN-38, the active metabolite of irinotecan. Irinotecan 72-82 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 9789606-1 1998 BACKGROUND: Irinotecan (CPT-11) is metabolized by esterase to form a SN-38, which is further conjugated by UGT1A1. Irinotecan 12-22 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 107-113 9789606-1 1998 BACKGROUND: Irinotecan (CPT-11) is metabolized by esterase to form a SN-38, which is further conjugated by UGT1A1. Irinotecan 24-30 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 107-113 9789606-1 1998 BACKGROUND: Irinotecan (CPT-11) is metabolized by esterase to form a SN-38, which is further conjugated by UGT1A1. Irinotecan 69-74 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 107-113 9789606-3 1998 We investigated whether there might be an inter-individual difference in pharmacokinetics of SN-38 and its glucuronide, depending on the genotypes of UGT1A1. Irinotecan 93-98 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 150-156 9569026-9 1998 Bax-K562 cells showed significantly higher fractions of apoptotic cells than pCI-neo-K562 cells when treated with ara-C, doxorubicin or SN-38. Irinotecan 136-141 BCL2 associated X, apoptosis regulator Homo sapiens 0-3 9472629-0 1998 CPT-11 sensitivity in relation to the expression of P170-glycoprotein and multidrug resistance-associated protein. Irinotecan 0-6 phosphoglycolate phosphatase Homo sapiens 52-69 9466980-5 1998 The purpose of this study was to identify the specific isoform of UGT involved in SN-38 glucuronidation. Irinotecan 82-87 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 66-69 9466980-8 1998 Gunn rats and CN-I patients lacked SN-38 glucuronidating activity, indicating the role of UGT1 isoform in SN-38 glucuronidation. Irinotecan 106-111 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 90-94 9466980-10 1998 Intact SN-38 glucuronidation was observed only in HK293 cells transfected with the UGT1A1 isozyme. Irinotecan 7-12 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 83-89 9466980-11 1998 These results demonstrate that UGT1A1 is the isoform responsible for SN-38 glucuronidation. Irinotecan 69-74 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 31-37 9466980-12 1998 These findings indicate a genetic predisposition to the metabolism of irinotecan, suggesting that patients with low UGT1A1 activity, such as those with Gilbert"s syndrome, may be at an increased risk for irinotecan toxicity. Irinotecan 70-80 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 116-122 9466980-12 1998 These findings indicate a genetic predisposition to the metabolism of irinotecan, suggesting that patients with low UGT1A1 activity, such as those with Gilbert"s syndrome, may be at an increased risk for irinotecan toxicity. Irinotecan 204-214 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 116-122 9533544-7 1998 In addition, we evaluated the effect of a sensitive (HC1) and resistant (ELC2) human colon adenocarcinoma xenograft on irinotecan and SN-38 lactone disposition after administration of irinotecan 10 mg/kg i.v. Irinotecan 119-129 CYCS pseudogene 39 Homo sapiens 53-56 9533544-7 1998 In addition, we evaluated the effect of a sensitive (HC1) and resistant (ELC2) human colon adenocarcinoma xenograft on irinotecan and SN-38 lactone disposition after administration of irinotecan 10 mg/kg i.v. Irinotecan 119-129 elaC ribonuclease Z 2 Homo sapiens 73-77 9533544-15 1998 administration, mice bearing HC1 xenografts had 43% greater SN-38 lactone systemic exposure compared to those with ELC2 xenografts and non-tumor-bearing mice. Irinotecan 60-73 heterochromatin, Chr 1 Mus musculus 29-32 9533544-18 1998 SN-38 systemic exposure associated with the lowest oral dose (25 mg/kg) achieving complete response for HC1 was 942.6 ng/ml x h. These results emphasize the importance of pharmacokinetic studies as part of tumor response studies in xenograft models. Irinotecan 0-5 CYCS pseudogene 39 Homo sapiens 104-107 9472629-0 1998 CPT-11 sensitivity in relation to the expression of P170-glycoprotein and multidrug resistance-associated protein. Irinotecan 0-6 ATP binding cassette subfamily C member 3 Homo sapiens 74-113 9472629-1 1998 The relevance of P170-glycoprotein (P-gp) and multidrug resistance-associated protein (MRP) for the sensitivity to CPT-11 was investigated in human malignant cell lines as well as in human tumour xenografts. Irinotecan 115-121 phosphoglycolate phosphatase Homo sapiens 17-34 9472629-1 1998 The relevance of P170-glycoprotein (P-gp) and multidrug resistance-associated protein (MRP) for the sensitivity to CPT-11 was investigated in human malignant cell lines as well as in human tumour xenografts. Irinotecan 115-121 phosphoglycolate phosphatase Homo sapiens 36-40 9472629-1 1998 The relevance of P170-glycoprotein (P-gp) and multidrug resistance-associated protein (MRP) for the sensitivity to CPT-11 was investigated in human malignant cell lines as well as in human tumour xenografts. Irinotecan 115-121 ATP binding cassette subfamily C member 3 Homo sapiens 46-85 9472629-1 1998 The relevance of P170-glycoprotein (P-gp) and multidrug resistance-associated protein (MRP) for the sensitivity to CPT-11 was investigated in human malignant cell lines as well as in human tumour xenografts. Irinotecan 115-121 ATP binding cassette subfamily C member 3 Homo sapiens 87-90 9472629-4 1998 The P-gp modulators BIBW22BS, verapamil and dexniguldipine partly reversed the resistance against CPT-11 in the P-gp-positive sublines. Irinotecan 98-104 phosphoglycolate phosphatase Homo sapiens 4-8 9472629-4 1998 The P-gp modulators BIBW22BS, verapamil and dexniguldipine partly reversed the resistance against CPT-11 in the P-gp-positive sublines. Irinotecan 98-104 phosphoglycolate phosphatase Homo sapiens 112-116 9472629-6 1998 In contrast to doxorubicin and vincristine, the BRO/mdr1.1 xenografts were at least as sensitive to CPT-11 as the BRO xenografts. Irinotecan 100-106 ATP binding cassette subfamily B member 1 Homo sapiens 52-56 9472629-9 1998 The MRP-positive subline GLC4/ADR was cross-resistant against carboxylesterase-activated CPT-11 and SN-38. Irinotecan 89-95 ATP binding cassette subfamily C member 3 Homo sapiens 4-7 9472629-9 1998 The MRP-positive subline GLC4/ADR was cross-resistant against carboxylesterase-activated CPT-11 and SN-38. Irinotecan 89-95 aldo-keto reductase family 1 member B Homo sapiens 30-33 9426050-0 1998 Overexpression of a rabbit liver carboxylesterase sensitizes human tumor cells to CPT-11. Irinotecan 82-88 liver carboxylesterase 1 Oryctolagus cuniculus 27-49 9472629-9 1998 The MRP-positive subline GLC4/ADR was cross-resistant against carboxylesterase-activated CPT-11 and SN-38. Irinotecan 100-105 ATP binding cassette subfamily C member 3 Homo sapiens 4-7 9426050-3 1998 Data in this study show metabolism of CPT-11 to SN-38 (7-ethyl-10-hydroxycamptothecin) by a rabbit liver carboxylesterase in vitro and growth-inhibitory activity of the products of the reaction. Irinotecan 38-44 liver carboxylesterase 1 Oryctolagus cuniculus 99-121 9426050-3 1998 Data in this study show metabolism of CPT-11 to SN-38 (7-ethyl-10-hydroxycamptothecin) by a rabbit liver carboxylesterase in vitro and growth-inhibitory activity of the products of the reaction. Irinotecan 48-53 liver carboxylesterase 1 Oryctolagus cuniculus 99-121 9426050-3 1998 Data in this study show metabolism of CPT-11 to SN-38 (7-ethyl-10-hydroxycamptothecin) by a rabbit liver carboxylesterase in vitro and growth-inhibitory activity of the products of the reaction. Irinotecan 55-85 liver carboxylesterase 1 Oryctolagus cuniculus 99-121 9744772-0 1998 Inhibition of intestinal microflora beta-glucuronidase modifies the distribution of the active metabolite of the antitumor agent, irinotecan hydrochloride (CPT-11) in rats. Irinotecan 130-154 glucuronidase, beta Rattus norvegicus 36-54 9472629-9 1998 The MRP-positive subline GLC4/ADR was cross-resistant against carboxylesterase-activated CPT-11 and SN-38. Irinotecan 100-105 aldo-keto reductase family 1 member B Homo sapiens 30-33 9744772-0 1998 Inhibition of intestinal microflora beta-glucuronidase modifies the distribution of the active metabolite of the antitumor agent, irinotecan hydrochloride (CPT-11) in rats. Irinotecan 156-162 glucuronidase, beta Rattus norvegicus 36-54 9393754-0 1997 Intravenous administration of irinotecan elevates the blood beta-glucuronidase activity in rats. Irinotecan 30-40 glucuronidase, beta Rattus norvegicus 60-78 9744772-2 1998 We have previously found that the inhibition of beta-glucuronidase, which hydrolyzes detoxified SN-38 (SN-38 glucuronide) to reform SN-38, in the lumen by eliminating the intestinal microflora with antibiotics, markedly ameliorates the intestinal toxicity of CPT-11 in rats. Irinotecan 96-101 glucuronidase, beta Rattus norvegicus 48-66 9744772-2 1998 We have previously found that the inhibition of beta-glucuronidase, which hydrolyzes detoxified SN-38 (SN-38 glucuronide) to reform SN-38, in the lumen by eliminating the intestinal microflora with antibiotics, markedly ameliorates the intestinal toxicity of CPT-11 in rats. Irinotecan 103-108 glucuronidase, beta Rattus norvegicus 48-66 9744772-2 1998 We have previously found that the inhibition of beta-glucuronidase, which hydrolyzes detoxified SN-38 (SN-38 glucuronide) to reform SN-38, in the lumen by eliminating the intestinal microflora with antibiotics, markedly ameliorates the intestinal toxicity of CPT-11 in rats. Irinotecan 259-265 glucuronidase, beta Rattus norvegicus 48-66 9744772-9 1998 CONCLUSIONS: These results suggest that SN-38, which results from the hydrolysis of SN-38 glucuronide by beta-glucuronidase in the intestinal microflora, contributes considerably to the distribution of SN-38 in the large intestine tissue, and that inhibition of the beta-glucuronidase activity by antibiotics results in decreased accumulation of SN-38 in the large intestine. Irinotecan 40-45 glucuronidase, beta Rattus norvegicus 105-123 9744772-9 1998 CONCLUSIONS: These results suggest that SN-38, which results from the hydrolysis of SN-38 glucuronide by beta-glucuronidase in the intestinal microflora, contributes considerably to the distribution of SN-38 in the large intestine tissue, and that inhibition of the beta-glucuronidase activity by antibiotics results in decreased accumulation of SN-38 in the large intestine. Irinotecan 40-45 glucuronidase, beta Rattus norvegicus 266-284 9744772-9 1998 CONCLUSIONS: These results suggest that SN-38, which results from the hydrolysis of SN-38 glucuronide by beta-glucuronidase in the intestinal microflora, contributes considerably to the distribution of SN-38 in the large intestine tissue, and that inhibition of the beta-glucuronidase activity by antibiotics results in decreased accumulation of SN-38 in the large intestine. Irinotecan 84-89 glucuronidase, beta Rattus norvegicus 105-123 9744772-9 1998 CONCLUSIONS: These results suggest that SN-38, which results from the hydrolysis of SN-38 glucuronide by beta-glucuronidase in the intestinal microflora, contributes considerably to the distribution of SN-38 in the large intestine tissue, and that inhibition of the beta-glucuronidase activity by antibiotics results in decreased accumulation of SN-38 in the large intestine. Irinotecan 84-89 glucuronidase, beta Rattus norvegicus 266-284 9744772-9 1998 CONCLUSIONS: These results suggest that SN-38, which results from the hydrolysis of SN-38 glucuronide by beta-glucuronidase in the intestinal microflora, contributes considerably to the distribution of SN-38 in the large intestine tissue, and that inhibition of the beta-glucuronidase activity by antibiotics results in decreased accumulation of SN-38 in the large intestine. Irinotecan 84-89 glucuronidase, beta Rattus norvegicus 105-123 9744772-9 1998 CONCLUSIONS: These results suggest that SN-38, which results from the hydrolysis of SN-38 glucuronide by beta-glucuronidase in the intestinal microflora, contributes considerably to the distribution of SN-38 in the large intestine tissue, and that inhibition of the beta-glucuronidase activity by antibiotics results in decreased accumulation of SN-38 in the large intestine. Irinotecan 84-89 glucuronidase, beta Rattus norvegicus 266-284 9750028-3 1998 In vivo pharmacokinetic studies revealed that the biliary excretion of the four anionic forms of CPT-11 and its metabolites was reduced in Eisai hyperbilirubinemic rats, which carry a mutation of the hepatic canalicular multispecific organic anion transporter (cMOAT) gene. Irinotecan 97-103 ATP binding cassette subfamily C member 2 Rattus norvegicus 208-259 9750028-3 1998 In vivo pharmacokinetic studies revealed that the biliary excretion of the four anionic forms of CPT-11 and its metabolites was reduced in Eisai hyperbilirubinemic rats, which carry a mutation of the hepatic canalicular multispecific organic anion transporter (cMOAT) gene. Irinotecan 97-103 ATP binding cassette subfamily C member 2 Rattus norvegicus 261-266 9750028-5 1998 Detailed analysis using isolated liver bile canalicular membrane vesicles to identify transport systems showed that cMOAT is responsible for biliary excretion of the low-affinity component of the carboxylate form of CPT-11 and the high-affinity component of both the lactone and carboxylate forms of SN-38 glucuronide. Irinotecan 216-222 ATP binding cassette subfamily C member 2 Rattus norvegicus 116-121 9750028-6 1998 The carboxylate form of SN-38 is transported by cMOAT alone. Irinotecan 24-29 ATP binding cassette subfamily C member 2 Rattus norvegicus 48-53 9750028-8 1998 In addition, ATP dependence in the uptake of CPT-11 by membrane vesicles obtained from a P-glycoprotein (P-gp)-overexpressing cell line was observed. Irinotecan 45-51 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 89-103 9750028-8 1998 In addition, ATP dependence in the uptake of CPT-11 by membrane vesicles obtained from a P-glycoprotein (P-gp)-overexpressing cell line was observed. Irinotecan 45-51 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 105-109 9750028-9 1998 Thus P-gp may be responsible for transport of the high-affinity component of the carboxylate form of CPT-11. Irinotecan 101-107 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 5-9 9654107-6 1998 Simultaneous exposure to paclitaxel and SN-38 for 24 h produced antagonistic (subadditive and protective) effects in the human lung cancer cell line A549, the breast cancer cell line MCF7, and the colon cancer cell line WiDr, and produced additive effects in the ovarian cancer cell line PA1. Irinotecan 40-45 PAXIP1 associated glutamate rich protein 1 Homo sapiens 288-291 9393754-4 1997 Prompted by the enzymological and structural similarity of CPT-11 to organophosphorus and carbamate insecticides, we studied the effect of CPT-11 on blood beta-GL activity in rats. Irinotecan 139-145 glucuronidase, beta Rattus norvegicus 155-162 9393754-6 1997 injection of CPT-11 in rats significantly elevated their plasma beta-GL activity (with phenolphthalein glucuronide as a substrate) at doses of 10 and 40 mg/kg, with peak activity observed 2-3 h after administration. Irinotecan 13-19 glucuronidase, beta Rattus norvegicus 64-71 9157988-1 1997 We have reported previously that a canalicular multispecific organic anion transporter (cMOAT) is responsible for the biliary excretion of carboxylate forms of irinotecan, 7-ethyl-10-[4-(1-piperidino)-1 piperidino] carbonyloxy camptothecin (CPT-11), its active metabolite SN-38, and glucuronide conjugate (SN38-Glu) and the lactone form of SN38-Glu in rats. Irinotecan 160-170 ATP binding cassette subfamily C member 2 Rattus norvegicus 35-86 9402181-2 1997 Preclinical studies showed that UGT*1.1 is the isozyme responsible for SN-38 glucuronidation. Irinotecan 71-76 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 32-39 9402181-3 1997 Patients with Gilbert"s syndrome have deficient UGT*1.1 activity, therefore may have an increased risk for related CPT-11 toxicity. Irinotecan 115-121 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 48-55 9334824-1 1997 The anti-tumor activity of irinotecan (CPT-11), a DNA-topoisomerase 1 inhibitor, was evaluated in 5 advanced stage subcutaneous medulloblastoma xenografts in nude mice, using different schedules of administration. Irinotecan 27-37 topoisomerase (DNA) I Mus musculus 50-69 9334824-1 1997 The anti-tumor activity of irinotecan (CPT-11), a DNA-topoisomerase 1 inhibitor, was evaluated in 5 advanced stage subcutaneous medulloblastoma xenografts in nude mice, using different schedules of administration. Irinotecan 39-45 topoisomerase (DNA) I Mus musculus 50-69 9473740-4 1997 Furthermore, sensitivity of PC-7/CPT cells to SN-38 was improved by inhibiting UGT activity. Irinotecan 46-51 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 79-82 9473740-6 1997 These results not only imply that upregulation of UGT activity in PC-7/CPT cells may contribute in part to SN-38 resistance, but also illustrate the important of drug metabolism within malignant cells themselves, as a cause of drug resistance. Irinotecan 107-112 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 50-53 9345343-0 1997 Irinotecan hydrochloride (CPT-11) resistance identified by K-ras mutation in patients with progressive colon cancer after treatment with 5-fluorouracil (5-FU). Irinotecan 0-24 KRAS proto-oncogene, GTPase Homo sapiens 59-64 9345343-0 1997 Irinotecan hydrochloride (CPT-11) resistance identified by K-ras mutation in patients with progressive colon cancer after treatment with 5-fluorouracil (5-FU). Irinotecan 26-32 KRAS proto-oncogene, GTPase Homo sapiens 59-64 9345343-1 1997 OBJECTIVE: To determine the prognostic role of a K-ras mutation in tumor tissue of patients with refractory colon cancer who received irinotecan hydrochloride (CPT-11). Irinotecan 134-158 KRAS proto-oncogene, GTPase Homo sapiens 49-54 9345343-1 1997 OBJECTIVE: To determine the prognostic role of a K-ras mutation in tumor tissue of patients with refractory colon cancer who received irinotecan hydrochloride (CPT-11). Irinotecan 160-166 KRAS proto-oncogene, GTPase Homo sapiens 49-54 9345343-9 1997 CONCLUSION: Determination of the presence of a K-ras mutation may predict survival in patients with progressive colon cancer after treatment with 5-fluorouracil who receive CPT-11. Irinotecan 173-179 KRAS proto-oncogene, GTPase Homo sapiens 47-52 9414664-4 1997 Ectopic p16INK4-expressing cells also showed approximately 4.0-fold increase in sensitivity to SN-38 (7-ethyl-10-hydroxycamptothecin), the active metabolite of CPT-11, compared to the parent cells. Irinotecan 95-100 cyclin dependent kinase inhibitor 2A Homo sapiens 8-15 9414664-4 1997 Ectopic p16INK4-expressing cells also showed approximately 4.0-fold increase in sensitivity to SN-38 (7-ethyl-10-hydroxycamptothecin), the active metabolite of CPT-11, compared to the parent cells. Irinotecan 102-132 cyclin dependent kinase inhibitor 2A Homo sapiens 8-15 9414664-4 1997 Ectopic p16INK4-expressing cells also showed approximately 4.0-fold increase in sensitivity to SN-38 (7-ethyl-10-hydroxycamptothecin), the active metabolite of CPT-11, compared to the parent cells. Irinotecan 160-166 cyclin dependent kinase inhibitor 2A Homo sapiens 8-15 9259410-3 1997 In this study, we established an SN-38-resistant cell line, PC-6/SN2-5, from the human oat cell carcinoma PC-6 cell line by a stepwise selection system, investigated the mechanism of resistance of this cell line and then compared the antitumor activity of camptothecin analogs against the cell line. Irinotecan 33-38 proprotein convertase subtilisin/kexin type 5 Homo sapiens 60-64 9259410-3 1997 In this study, we established an SN-38-resistant cell line, PC-6/SN2-5, from the human oat cell carcinoma PC-6 cell line by a stepwise selection system, investigated the mechanism of resistance of this cell line and then compared the antitumor activity of camptothecin analogs against the cell line. Irinotecan 33-38 proprotein convertase subtilisin/kexin type 5 Homo sapiens 106-110 9259410-4 1997 PC-6/SN2-5 cells were resistant to SN-38 (32-fold) and SK&F 104864 (topotecan; 14-fold), but barely resistant to CPT-11 (3-fold) and DX-8951f (2-fold). Irinotecan 35-40 proprotein convertase subtilisin/kexin type 5 Homo sapiens 0-4 9259410-4 1997 PC-6/SN2-5 cells were resistant to SN-38 (32-fold) and SK&F 104864 (topotecan; 14-fold), but barely resistant to CPT-11 (3-fold) and DX-8951f (2-fold). Irinotecan 117-123 proprotein convertase subtilisin/kexin type 5 Homo sapiens 0-4 9259410-6 1997 Determination of the cellular drug concentration by either flow cytometric analysis or the high-performance liquid chromatography method confirmed that the cellular accumulation of SN-38 and topotecan was significantly reduced in PC-6/SN2-5 cells, whereas that of DX-8951f was only slightly reduced. Irinotecan 181-186 proprotein convertase subtilisin/kexin type 5 Homo sapiens 230-234 9184074-0 1997 Sequential operation of ceramide synthesis and ICE cascade in CPT-11-initiated apoptotic death signaling. Irinotecan 62-68 caspase 1 Mus musculus 47-50 9184074-3 1997 CPT-11-initiated cytolytic activity was prevented by both caspase inhibitors YVAD-CHO and DEVD-CHO, or ceramide synthesis inhibitor fumonisin B1, and accelerated by sphingomyelin, suggesting the direct involvement of ceramide synthesis and the interleukin 1-beta converting enzyme (ICE) cascade. Irinotecan 0-6 caspase 1 Mus musculus 58-65 9184074-3 1997 CPT-11-initiated cytolytic activity was prevented by both caspase inhibitors YVAD-CHO and DEVD-CHO, or ceramide synthesis inhibitor fumonisin B1, and accelerated by sphingomyelin, suggesting the direct involvement of ceramide synthesis and the interleukin 1-beta converting enzyme (ICE) cascade. Irinotecan 0-6 caspase 1 Mus musculus 244-280 9184074-3 1997 CPT-11-initiated cytolytic activity was prevented by both caspase inhibitors YVAD-CHO and DEVD-CHO, or ceramide synthesis inhibitor fumonisin B1, and accelerated by sphingomyelin, suggesting the direct involvement of ceramide synthesis and the interleukin 1-beta converting enzyme (ICE) cascade. Irinotecan 0-6 caspase 1 Mus musculus 282-285 9157988-1 1997 We have reported previously that a canalicular multispecific organic anion transporter (cMOAT) is responsible for the biliary excretion of carboxylate forms of irinotecan, 7-ethyl-10-[4-(1-piperidino)-1 piperidino] carbonyloxy camptothecin (CPT-11), its active metabolite SN-38, and glucuronide conjugate (SN38-Glu) and the lactone form of SN38-Glu in rats. Irinotecan 241-247 ATP binding cassette subfamily C member 2 Rattus norvegicus 35-86 9157988-1 1997 We have reported previously that a canalicular multispecific organic anion transporter (cMOAT) is responsible for the biliary excretion of carboxylate forms of irinotecan, 7-ethyl-10-[4-(1-piperidino)-1 piperidino] carbonyloxy camptothecin (CPT-11), its active metabolite SN-38, and glucuronide conjugate (SN38-Glu) and the lactone form of SN38-Glu in rats. Irinotecan 241-247 ATP binding cassette subfamily C member 2 Rattus norvegicus 88-93 9157988-1 1997 We have reported previously that a canalicular multispecific organic anion transporter (cMOAT) is responsible for the biliary excretion of carboxylate forms of irinotecan, 7-ethyl-10-[4-(1-piperidino)-1 piperidino] carbonyloxy camptothecin (CPT-11), its active metabolite SN-38, and glucuronide conjugate (SN38-Glu) and the lactone form of SN38-Glu in rats. Irinotecan 272-277 ATP binding cassette subfamily C member 2 Rattus norvegicus 35-86 9157988-1 1997 We have reported previously that a canalicular multispecific organic anion transporter (cMOAT) is responsible for the biliary excretion of carboxylate forms of irinotecan, 7-ethyl-10-[4-(1-piperidino)-1 piperidino] carbonyloxy camptothecin (CPT-11), its active metabolite SN-38, and glucuronide conjugate (SN38-Glu) and the lactone form of SN38-Glu in rats. Irinotecan 272-277 ATP binding cassette subfamily C member 2 Rattus norvegicus 88-93 9157988-5 1997 The carboxylate form of SN-38 was found to be transported by cMOAT alone. Irinotecan 24-29 ATP binding cassette subfamily C member 2 Rattus norvegicus 61-66 9157988-6 1997 We conclude that multiple transporters, including cMOAT, are responsible for the biliary excretion of CPT-11 and its metabolites (anionic forms), and the contribution of each transporter differs greatly, depending on the substrates. Irinotecan 102-108 ATP binding cassette subfamily C member 2 Rattus norvegicus 50-55 9157988-1 1997 We have reported previously that a canalicular multispecific organic anion transporter (cMOAT) is responsible for the biliary excretion of carboxylate forms of irinotecan, 7-ethyl-10-[4-(1-piperidino)-1 piperidino] carbonyloxy camptothecin (CPT-11), its active metabolite SN-38, and glucuronide conjugate (SN38-Glu) and the lactone form of SN38-Glu in rats. Irinotecan 160-170 ATP binding cassette subfamily C member 2 Rattus norvegicus 88-93 8891470-2 1996 Irinotecan (CPT-II) is a semisynthetic derivative of camptothecin. Irinotecan 0-10 carnitine palmitoyltransferase 2 Homo sapiens 12-18 9095327-3 1997 In the SCLC cell line, supra-additive effect was observed for SN-38 in combination with cisplatin, etoposide (VP-16) and paclitaxel (Taxol). Irinotecan 62-67 host cell factor C1 Homo sapiens 110-115 20650253-0 1996 Involvement of CPP32/Yama-like protease in CPT-11-induced death signal transduction pathway. Irinotecan 43-49 caspase 3 Homo sapiens 15-20 20650253-0 1996 Involvement of CPP32/Yama-like protease in CPT-11-induced death signal transduction pathway. Irinotecan 43-49 caspase 3 Homo sapiens 21-25 20650253-1 1996 CPT-11, a derivative of camptothecin (CPT) that interacts with type-I DNA topoisomerase, induced apoptosis in HL60 and Daudi cells in vitro. Irinotecan 0-6 DNA topoisomerase I Homo sapiens 63-87 8922198-1 1996 BACKGROUND: CPT-11 (irinotecan), a camptothecin-derived anticancer agent with DNA topoisomerase 1 inhibitory activity, has demonstrated a broad spectrum of in vitro and in vivo activity in solid tumour models including multidrug-resistant tumours. Irinotecan 12-18 DNA topoisomerase I Homo sapiens 78-97 8922198-1 1996 BACKGROUND: CPT-11 (irinotecan), a camptothecin-derived anticancer agent with DNA topoisomerase 1 inhibitory activity, has demonstrated a broad spectrum of in vitro and in vivo activity in solid tumour models including multidrug-resistant tumours. Irinotecan 20-30 DNA topoisomerase I Homo sapiens 78-97 9095327-9 1997 These results suggest that all the drugs we selected can be used with SN-38 simultaneously for NSCLC, while for SCLC, cisplatin, VP-16 and Taxol are the most suitable for combination with SN-38. Irinotecan 188-193 host cell factor C1 Homo sapiens 129-134 9155540-0 1997 A phase I study of irinotecan and infusional cisplatin with recombinant human granulocyte colony-stimulating factor support in the treatment of advanced non-small cell lung cancer. Irinotecan 19-29 colony stimulating factor 3 Homo sapiens 78-115 9155540-1 1997 We conducted a phase I study to examine whether support with recombinant human granulocyte colony-stimulating factor (rG-CSF) would permit dose intensification of irinotecan (CPT-11) in combination with cisplatin (20 mg/m2 x 5 days) in non-small cell lung cancer (NSCLC) patients. Irinotecan 163-173 colony stimulating factor 3 Homo sapiens 79-116 9155540-1 1997 We conducted a phase I study to examine whether support with recombinant human granulocyte colony-stimulating factor (rG-CSF) would permit dose intensification of irinotecan (CPT-11) in combination with cisplatin (20 mg/m2 x 5 days) in non-small cell lung cancer (NSCLC) patients. Irinotecan 163-173 colony stimulating factor 3 Rattus norvegicus 118-124 9155540-1 1997 We conducted a phase I study to examine whether support with recombinant human granulocyte colony-stimulating factor (rG-CSF) would permit dose intensification of irinotecan (CPT-11) in combination with cisplatin (20 mg/m2 x 5 days) in non-small cell lung cancer (NSCLC) patients. Irinotecan 175-181 colony stimulating factor 3 Homo sapiens 79-116 9155540-1 1997 We conducted a phase I study to examine whether support with recombinant human granulocyte colony-stimulating factor (rG-CSF) would permit dose intensification of irinotecan (CPT-11) in combination with cisplatin (20 mg/m2 x 5 days) in non-small cell lung cancer (NSCLC) patients. Irinotecan 175-181 colony stimulating factor 3 Rattus norvegicus 118-124 9033637-10 1997 We observed a positive relationship between the DNA topoisomerase-1 activity and the cellular sensitivity to carboxylesterase-activated CPT-11 (r = 0.75, p < 0.1) as well as to SN-38 (r = 0.89, p < 0.05). Irinotecan 136-142 DNA topoisomerase I Homo sapiens 48-67 9033637-10 1997 We observed a positive relationship between the DNA topoisomerase-1 activity and the cellular sensitivity to carboxylesterase-activated CPT-11 (r = 0.75, p < 0.1) as well as to SN-38 (r = 0.89, p < 0.05). Irinotecan 180-185 DNA topoisomerase I Homo sapiens 48-67 9033637-12 1997 In conclusion, the DNA topoisomerase-1 activity was the best determinant for CPT-11/SN-38 sensitivity in this panel of unselected human colon-cancer cell lines. Irinotecan 77-83 DNA topoisomerase I Homo sapiens 19-38 9033637-12 1997 In conclusion, the DNA topoisomerase-1 activity was the best determinant for CPT-11/SN-38 sensitivity in this panel of unselected human colon-cancer cell lines. Irinotecan 84-89 DNA topoisomerase I Homo sapiens 19-38 9054958-1 1997 PURPOSE: Irinotecan (CPT-11) is hydrolyzed to its active metabolite SN-38 which is subsequently conjugated by uridine diphosphate glucuronosyl transferase (UDP-GT) to the glucuronide (SN-38G). Irinotecan 9-19 UDP glucuronosyltransferase family 1 member A1 Rattus norvegicus 110-154 9054958-1 1997 PURPOSE: Irinotecan (CPT-11) is hydrolyzed to its active metabolite SN-38 which is subsequently conjugated by uridine diphosphate glucuronosyl transferase (UDP-GT) to the glucuronide (SN-38G). Irinotecan 9-19 UDP glucuronosyltransferase family 1 member A1 Rattus norvegicus 156-162 9054958-1 1997 PURPOSE: Irinotecan (CPT-11) is hydrolyzed to its active metabolite SN-38 which is subsequently conjugated by uridine diphosphate glucuronosyl transferase (UDP-GT) to the glucuronide (SN-38G). Irinotecan 21-27 UDP glucuronosyltransferase family 1 member A1 Rattus norvegicus 110-154 9054958-1 1997 PURPOSE: Irinotecan (CPT-11) is hydrolyzed to its active metabolite SN-38 which is subsequently conjugated by uridine diphosphate glucuronosyl transferase (UDP-GT) to the glucuronide (SN-38G). Irinotecan 21-27 UDP glucuronosyltransferase family 1 member A1 Rattus norvegicus 156-162 9054958-1 1997 PURPOSE: Irinotecan (CPT-11) is hydrolyzed to its active metabolite SN-38 which is subsequently conjugated by uridine diphosphate glucuronosyl transferase (UDP-GT) to the glucuronide (SN-38G). Irinotecan 68-73 UDP glucuronosyltransferase family 1 member A1 Rattus norvegicus 110-154 9054958-1 1997 PURPOSE: Irinotecan (CPT-11) is hydrolyzed to its active metabolite SN-38 which is subsequently conjugated by uridine diphosphate glucuronosyl transferase (UDP-GT) to the glucuronide (SN-38G). Irinotecan 68-73 UDP glucuronosyltransferase family 1 member A1 Rattus norvegicus 156-162 9054958-1 1997 PURPOSE: Irinotecan (CPT-11) is hydrolyzed to its active metabolite SN-38 which is subsequently conjugated by uridine diphosphate glucuronosyl transferase (UDP-GT) to the glucuronide (SN-38G). Irinotecan 184-189 UDP glucuronosyltransferase family 1 member A1 Rattus norvegicus 110-154 9054958-1 1997 PURPOSE: Irinotecan (CPT-11) is hydrolyzed to its active metabolite SN-38 which is subsequently conjugated by uridine diphosphate glucuronosyl transferase (UDP-GT) to the glucuronide (SN-38G). Irinotecan 184-189 UDP glucuronosyltransferase family 1 member A1 Rattus norvegicus 156-162 9030234-4 1997 Introduction of bcl 2 gene into human small cell lung cancer cells conferred resistance to mitomycin C and irinotecan. Irinotecan 107-117 BCL2 apoptosis regulator Homo sapiens 16-21 9121221-0 1996 Antitumor effect of CPT-11, a new derivative of camptothecin, against human prostate cancer (PC-3) in vitro and prostate rat tumor (AT-3) in vivo. Irinotecan 20-26 chromobox 8 Homo sapiens 76-97 9121221-4 1996 CPT-11 at concentrations between 10 ng/ml and 50 micrograms/ml inhibited the growth of the PC-3 prostate human cancer cell line up < 0.001). Irinotecan 0-6 chromobox 8 Homo sapiens 91-95 20650253-8 1996 These results indicate the direct involvement of CPP32/Yama-like protease in the CPT-11-induced death signal transduction pathway, and no involvement of Fas antigen in the pathway. Irinotecan 81-87 caspase 3 Homo sapiens 49-54 20650253-8 1996 These results indicate the direct involvement of CPP32/Yama-like protease in the CPT-11-induced death signal transduction pathway, and no involvement of Fas antigen in the pathway. Irinotecan 81-87 caspase 3 Homo sapiens 55-59 8917343-0 1996 Induction of tumor necrosis factor by a camptothecin derivative, irinotecan, in mice and human mononuclear cells. Irinotecan 65-75 tumor necrosis factor Mus musculus 13-34 8917343-1 1996 Irinotecan (CPT-11) has been reported to be cytotoxic to tumor cells through its inhibitory activity on type I DNA topoisomerase. Irinotecan 0-10 DNA topoisomerase I Homo sapiens 104-128 8917343-1 1996 Irinotecan (CPT-11) has been reported to be cytotoxic to tumor cells through its inhibitory activity on type I DNA topoisomerase. Irinotecan 12-18 DNA topoisomerase I Homo sapiens 104-128 8917343-3 1996 We investigated the ability of CPT-11 to induce tumor necrosis factor (TNF) production. Irinotecan 31-37 tumor necrosis factor Homo sapiens 48-69 8917343-3 1996 We investigated the ability of CPT-11 to induce tumor necrosis factor (TNF) production. Irinotecan 31-37 tumor necrosis factor Homo sapiens 71-74 8917343-5 1996 The priming effect of CPT-11 on endogenous production of TNF was examined by injecting the drug intravenously into mice, followed 3 hours by the injection of OK432. Irinotecan 22-28 tumor necrosis factor Mus musculus 57-60 8917343-7 1996 A significant amount of TNF was released when MNCs were incubated with 100-300 microM of CPT-11 for more than 4 hours, but not with vinblastin sulfate, indicating a triggering effect of TNF production on MNCs in vitro. Irinotecan 89-95 tumor necrosis factor Homo sapiens 24-27 8917343-7 1996 A significant amount of TNF was released when MNCs were incubated with 100-300 microM of CPT-11 for more than 4 hours, but not with vinblastin sulfate, indicating a triggering effect of TNF production on MNCs in vitro. Irinotecan 89-95 tumor necrosis factor Homo sapiens 186-189 8706020-0 1996 Involvement of beta-glucuronidase in intestinal microflora in the intestinal toxicity of the antitumor camptothecin derivative irinotecan hydrochloride (CPT-11) in rats. Irinotecan 127-151 glucuronidase, beta Rattus norvegicus 15-33 8841050-16 1996 In conclusion, adverse reactions caused by the combination therapy with CPT-11 and CDDP (CPT-11: 50-60 mg/m2 on days 1, 8 and 15, CDDP: 50-60 mg/m2 on day 1) can be relieved by short term administration of G-CSF and it is suggested that the combination therapy may be effective in treating ovarian carcinoma. Irinotecan 72-78 colony stimulating factor 3 Homo sapiens 206-211 8841050-16 1996 In conclusion, adverse reactions caused by the combination therapy with CPT-11 and CDDP (CPT-11: 50-60 mg/m2 on days 1, 8 and 15, CDDP: 50-60 mg/m2 on day 1) can be relieved by short term administration of G-CSF and it is suggested that the combination therapy may be effective in treating ovarian carcinoma. Irinotecan 89-95 colony stimulating factor 3 Homo sapiens 206-211 8706020-0 1996 Involvement of beta-glucuronidase in intestinal microflora in the intestinal toxicity of the antitumor camptothecin derivative irinotecan hydrochloride (CPT-11) in rats. Irinotecan 153-159 glucuronidase, beta Rattus norvegicus 15-33 8706020-8 1996 Analysis of CPT-11 and its metabolites in the feces indicated that antibiotics completely inhibited the deconjugation of the glucuronic conjugate of 7-ethyl-10-hydroxycamptothecin by beta-glucuronidase. Irinotecan 12-18 glucuronidase, beta Rattus norvegicus 183-201 8706020-8 1996 Analysis of CPT-11 and its metabolites in the feces indicated that antibiotics completely inhibited the deconjugation of the glucuronic conjugate of 7-ethyl-10-hydroxycamptothecin by beta-glucuronidase. Irinotecan 149-179 glucuronidase, beta Rattus norvegicus 183-201 8706020-9 1996 It is suggested that CPT-11-induced diarrhea would be attributable to the damage to the cecum, and that the inhibition of the beta-glucuronidase activity in the intestinal microflora is a major protective effect of antibiotics. Irinotecan 21-27 glucuronidase, beta Rattus norvegicus 126-144 8625310-11 1996 Treatment with CPT-11 or SN-38 also induced a markedly greater and more persistent reduction in topo I mRNA abundance in KB/STP-2 cells than in KB cells. Irinotecan 15-21 sulfotransferase family 1A member 2 Homo sapiens 124-129 8683231-2 1996 MATERIALS AND METHODS: The recommended phase II dose of weekly CPT-11 administered as a 90-minute infusion is 145 mg/m2 with granulocyte colony-stimulating factor (G-CSF) and maximal antidiarrheal support. Irinotecan 63-69 colony stimulating factor 3 Homo sapiens 125-162 8683231-2 1996 MATERIALS AND METHODS: The recommended phase II dose of weekly CPT-11 administered as a 90-minute infusion is 145 mg/m2 with granulocyte colony-stimulating factor (G-CSF) and maximal antidiarrheal support. Irinotecan 63-69 colony stimulating factor 3 Homo sapiens 164-169 8625310-11 1996 Treatment with CPT-11 or SN-38 also induced a markedly greater and more persistent reduction in topo I mRNA abundance in KB/STP-2 cells than in KB cells. Irinotecan 25-30 sulfotransferase family 1A member 2 Homo sapiens 124-129 8625310-14 1996 This novel regulation of topo I mRNA by topo I-targeting agents could be associated with acquirement of drug resistance to saintopin or SN-38/CPT-11 in KB/STP-2 cells. Irinotecan 136-141 sulfotransferase family 1A member 2 Homo sapiens 155-160 8625310-14 1996 This novel regulation of topo I mRNA by topo I-targeting agents could be associated with acquirement of drug resistance to saintopin or SN-38/CPT-11 in KB/STP-2 cells. Irinotecan 142-148 sulfotransferase family 1A member 2 Homo sapiens 155-160 8633248-3 1996 The antitumor effects of CPT-11 and SN-38 are exerted through a novel mechanism of action; inhibition of DNA topoisomerase I. Irinotecan 25-31 topoisomerase (DNA) I Mus musculus 105-124 8812723-0 1996 Stimulation of interleukin-1beta-converting enzyme activity during growth inhibition by CPT-11 in the human myeloid leukemia cell line K562. Irinotecan 88-94 caspase 1 Homo sapiens 15-50 8812723-6 1996 To determine how CPT-11 brings about cell death by apoptosis, we investigated the effects of CPT-11 on the expression of ICE activity in K562 cells, which represent human myeloid leukemia cells. Irinotecan 93-99 caspase 1 Homo sapiens 121-124 8812723-8 1996 We also found that the levels of mRNA for ICE in the cells were increased in the presence of CPT-11. Irinotecan 93-99 caspase 1 Homo sapiens 42-45 8812723-10 1996 These features suggested that CPT-11 enhances the apoptotic cell death in K562 cells and that a part of induction of apoptosis by CPT-11 may be correlated with the stimulation of the ICE activity. Irinotecan 30-36 caspase 1 Homo sapiens 183-186 8812723-10 1996 These features suggested that CPT-11 enhances the apoptotic cell death in K562 cells and that a part of induction of apoptosis by CPT-11 may be correlated with the stimulation of the ICE activity. Irinotecan 130-136 caspase 1 Homo sapiens 183-186 8633248-3 1996 The antitumor effects of CPT-11 and SN-38 are exerted through a novel mechanism of action; inhibition of DNA topoisomerase I. Irinotecan 36-41 topoisomerase (DNA) I Mus musculus 105-124 7733646-1 1995 The interaction between hyperthermia and a DNA topoisomerase I inhibitor, 7-ethyl-10-(4-(1-piperidyl)-1-piperidyl)-carbonyloxy- camptothecin (CPT-11), was studied in the mouse mammary carcinoma FM3A cells. Irinotecan 142-148 topoisomerase (DNA) I Mus musculus 43-62 7494232-1 1995 BACKGROUND: Irinotecan--or CPT-11; 7-ethyl-10-[4-(1-piperidino)-1-piperidino]-carbonyloxy-camptotheci n--is an inhibitor of DNA topoisomerase I and is clinically effective against several cancers. Irinotecan 12-22 topoisomerase (DNA) I Mus musculus 124-143 7494232-1 1995 BACKGROUND: Irinotecan--or CPT-11; 7-ethyl-10-[4-(1-piperidino)-1-piperidino]-carbonyloxy-camptotheci n--is an inhibitor of DNA topoisomerase I and is clinically effective against several cancers. Irinotecan 27-33 topoisomerase (DNA) I Mus musculus 124-143 7494232-1 1995 BACKGROUND: Irinotecan--or CPT-11; 7-ethyl-10-[4-(1-piperidino)-1-piperidino]-carbonyloxy-camptotheci n--is an inhibitor of DNA topoisomerase I and is clinically effective against several cancers. Irinotecan 35-105 topoisomerase (DNA) I Mus musculus 124-143 8664184-4 1995 Of those currently under investigation, CPT-11 seems to represent the most promising, since its mode of action is something other than inhibition of thymidylate synthase. Irinotecan 40-46 thymidylate synthetase Homo sapiens 149-169 7493918-2 1995 We investigated whether baicalin, an inhibitor of beta-glucuronidase, which deconjugates the glucuronide of the active metabolite of CPT-11, SN-38 (7-ethyl-10-hydorxycamptothecin), and Japanese herbal medicines (Kampo medicines) which contain baicalin can ameliorate CPT-11-induced intestinal toxicity in rats. Irinotecan 133-139 glucuronidase, beta Rattus norvegicus 50-68 7493918-2 1995 We investigated whether baicalin, an inhibitor of beta-glucuronidase, which deconjugates the glucuronide of the active metabolite of CPT-11, SN-38 (7-ethyl-10-hydorxycamptothecin), and Japanese herbal medicines (Kampo medicines) which contain baicalin can ameliorate CPT-11-induced intestinal toxicity in rats. Irinotecan 141-146 glucuronidase, beta Rattus norvegicus 50-68 7493918-2 1995 We investigated whether baicalin, an inhibitor of beta-glucuronidase, which deconjugates the glucuronide of the active metabolite of CPT-11, SN-38 (7-ethyl-10-hydorxycamptothecin), and Japanese herbal medicines (Kampo medicines) which contain baicalin can ameliorate CPT-11-induced intestinal toxicity in rats. Irinotecan 267-273 glucuronidase, beta Rattus norvegicus 50-68 7493919-0 1995 Inhibition of UDP-glucuronosyltransferase by aglycons of natural glucuronides in kampo medicines using SN-38 as a substrate. Irinotecan 103-108 UDP glucuronosyltransferase family 1 member A6 Homo sapiens 14-41 7493919-1 1995 7-Ethyl-10-[4-(piperidino)-1-piperidino]carbonyloxycamptothecin (CPT-11), a potent anticancer agent for lung and gynecological cancers, is metabolized in vivo to the active compound, 7-ethyl-10-hydroxycamptothecin (SN-38), which is subsequently conjugated to SN-38-glucuronide by UDP-glucuronosyltransferase (UDP-GT). Irinotecan 65-71 UDP glucuronosyltransferase family 1 member A6 Homo sapiens 280-307 7493919-1 1995 7-Ethyl-10-[4-(piperidino)-1-piperidino]carbonyloxycamptothecin (CPT-11), a potent anticancer agent for lung and gynecological cancers, is metabolized in vivo to the active compound, 7-ethyl-10-hydroxycamptothecin (SN-38), which is subsequently conjugated to SN-38-glucuronide by UDP-glucuronosyltransferase (UDP-GT). Irinotecan 65-71 UDP glucuronosyltransferase family 1 member A6 Homo sapiens 309-315 7493919-2 1995 Three purified aglycons of natural glucuronides, baicalein, luteolin and glycyrrhetic acid, inhibited UDP-GT activity towards SN-38 as a substrate. Irinotecan 126-131 UDP glucuronosyltransferase family 1 member A6 Homo sapiens 102-108 7766631-10 1995 The stability of 10-hydroxycamptothecin-induced cleavable complexes was intermediate to those of CPT and SN-38, indicating that both the 10-hydroxy and the 7-ethyl group of SN-38 probably interact with the drug binding site of top1-cleavable complexes. Irinotecan 173-178 choline phosphotransferase 1 Homo sapiens 97-100 7763013-0 1995 Sequence-dependent modulation of anticancer drug activities by 7-ethyl-10-hydroxycamptothecin in an HST-1 human squamous carcinoma cell line. Irinotecan 63-93 fibroblast growth factor 4 Homo sapiens 100-105 7763013-3 1995 RESULTS: SN-38 potentiated the antitumor activity of CDDP in all schedules with a maximal effect observed with a simultaneous administration, while SN-38 enhanced the cytotoxicity of MMC, 5-FU, or VP-16 only in certain schedules. Irinotecan 148-153 host cell factor C1 Homo sapiens 197-202 7763013-5 1995 CONCLUSIONS: These results demonstrate that SN-38 may have the advantage of augmenting the anticancer activity in combination with CDDP, MMC, 5-FU, and VP-16, depending on the schedule of administration, and should thus be incorporated into the design of a clinical trial for obtaining maximal therapeutic synergy. Irinotecan 44-49 host cell factor C1 Homo sapiens 152-157 7623066-6 1995 Total glutathione S-transferase (GST) and GST-p activities were similar in CPT-11-sensitive and -resistant cells. Irinotecan 75-81 glutathione S-transferase kappa 1 Homo sapiens 6-31 7623066-6 1995 Total glutathione S-transferase (GST) and GST-p activities were similar in CPT-11-sensitive and -resistant cells. Irinotecan 75-81 glutathione S-transferase kappa 1 Homo sapiens 33-36 7623066-6 1995 Total glutathione S-transferase (GST) and GST-p activities were similar in CPT-11-sensitive and -resistant cells. Irinotecan 75-81 glutathione S-transferase pi 1 Homo sapiens 42-47 7521905-0 1994 Phase I and pharmacologic study of irinotecan and etoposide with recombinant human granulocyte colony-stimulating factor support for advanced lung cancer. Irinotecan 35-45 colony stimulating factor 3 Homo sapiens 83-120 7737904-3 1995 However, HAC2/0.1, a CDDP-resistant human ovarian cancer cell line which is also resistant to CPT-11 because of decreased intracellular activation of CPT-11, was about 12.8-fold more resistant to KW-2189. Irinotecan 94-100 hyperpolarization activated cyclic nucleotide gated potassium channel 1 Homo sapiens 9-17 7737904-3 1995 However, HAC2/0.1, a CDDP-resistant human ovarian cancer cell line which is also resistant to CPT-11 because of decreased intracellular activation of CPT-11, was about 12.8-fold more resistant to KW-2189. Irinotecan 150-156 hyperpolarization activated cyclic nucleotide gated potassium channel 1 Homo sapiens 9-17 7737904-12 1995 The decreased conversion of CPT-11 to SN-38 in HAC2/0.1 cells might be explained by decreased carboxyl esterase activity. Irinotecan 28-34 hyperpolarization activated cyclic nucleotide gated potassium channel 1 Homo sapiens 47-51 7737904-12 1995 The decreased conversion of CPT-11 to SN-38 in HAC2/0.1 cells might be explained by decreased carboxyl esterase activity. Irinotecan 38-43 hyperpolarization activated cyclic nucleotide gated potassium channel 1 Homo sapiens 47-51 8074481-3 1994 An in vitro drug sensitivity assay revealed that HAC2/CPT was 9.7 and 4.7 times as resistant as HAC2 to CPT-11 and 7-ethyl-10-hydroxy-CPT-11 (SN-38), an active metabolite of CPT-11, respectively. Irinotecan 142-147 hyperpolarization activated cyclic nucleotide gated potassium channel 1 Homo sapiens 49-53 8060137-1 1994 In order to select a suitable combination chemotherapy with BOF-A2 from the view of both anti-tumor effect (IR) and decrease of side effect, we studied a combination significance of BOF-A2 with CPT-11 that promised for a new anticancer drug, CDDP or mitomycin C (MMC) that used widely to many cancer patients and interferon-alpha (IFN-alpha) against colon, stomach and renal cancer, respectively, by using xenografted nude mice. Irinotecan 194-200 interferon alpha 1 Homo sapiens 331-340 8089365-2 1994 In the present experiment, the possible modifying effects of Peplomycin (PEP, 30 mg/kg body weight) and Camptothecin-11, an inhibitor of DNA topoisomerase I (CPT-11, 15 and 50 mg/kg body weight), on radiation skin injury were studied. Irinotecan 104-119 topoisomerase (DNA) I Mus musculus 137-156 8074481-3 1994 An in vitro drug sensitivity assay revealed that HAC2/CPT was 9.7 and 4.7 times as resistant as HAC2 to CPT-11 and 7-ethyl-10-hydroxy-CPT-11 (SN-38), an active metabolite of CPT-11, respectively. Irinotecan 142-147 choline phosphotransferase 1 Homo sapiens 54-57 7920428-5 1994 In studies on the kinetic parameters of the hydrolysis of CPT-11 by the purified carboxylesterase isozymes the highest Vmax value of the isozymes was found in human HU1 and the smallest was seen in rat RL1. Irinotecan 58-64 homeobox B5 Homo sapiens 165-168 8074481-3 1994 An in vitro drug sensitivity assay revealed that HAC2/CPT was 9.7 and 4.7 times as resistant as HAC2 to CPT-11 and 7-ethyl-10-hydroxy-CPT-11 (SN-38), an active metabolite of CPT-11, respectively. Irinotecan 142-147 hyperpolarization activated cyclic nucleotide gated potassium channel 1 Homo sapiens 96-100 7920428-5 1994 In studies on the kinetic parameters of the hydrolysis of CPT-11 by the purified carboxylesterase isozymes the highest Vmax value of the isozymes was found in human HU1 and the smallest was seen in rat RL1. Irinotecan 58-64 ribonuclease A family member 1, pancreatic Rattus norvegicus 202-205 7505810-0 1994 Phase I study of irinotecan and cisplatin with granulocyte colony-stimulating factor support for advanced non-small-cell lung cancer. Irinotecan 17-27 colony stimulating factor 3 Homo sapiens 47-84 21566972-0 1994 Bcl-2 gene prevents induction of apoptosis in l1210 murine leukemia-cells by sn-38, a metabolite of the camptothecin derivative cpt-11. Irinotecan 77-82 B cell leukemia/lymphoma 2 Mus musculus 0-5 21566972-0 1994 Bcl-2 gene prevents induction of apoptosis in l1210 murine leukemia-cells by sn-38, a metabolite of the camptothecin derivative cpt-11. Irinotecan 128-134 B cell leukemia/lymphoma 2 Mus musculus 0-5 21566972-5 1994 We demonstrated in addition that enforced expression of the bcl-2 gene in L1210 cells by MPZenNeo (bcl-2) retroviral gene transfer increased resistance to the apoptosis induced by SN-38 and CPT. Irinotecan 180-185 B cell leukemia/lymphoma 2 Mus musculus 60-65 21566972-5 1994 We demonstrated in addition that enforced expression of the bcl-2 gene in L1210 cells by MPZenNeo (bcl-2) retroviral gene transfer increased resistance to the apoptosis induced by SN-38 and CPT. Irinotecan 180-185 B cell leukemia/lymphoma 2 Mus musculus 99-104 21566972-6 1994 These findings suggest the possibility that the bcl-2 gene impedes the activity of a common pathway for apoptosis induced by SN-38 and CPT. Irinotecan 125-130 B cell leukemia/lymphoma 2 Mus musculus 48-53 8070019-1 1994 The camptothecin derivative 7-ethyl-10-[4-(1-piperidino)-1-piperidino]-carbonyloxy camptothecin (CPT-11) has attracted the attention of clinicians because of its high antitumor activity against refractory solid cancers. Irinotecan 28-95 choline phosphotransferase 1 Homo sapiens 97-100 8070019-3 1994 The mechanisms of resistance to CPT-11 in PC-7/CPT-11 cells were reduced conversion of CPT-11 to its active metabolite SN-38 and point mutation topoisomerase I. Irinotecan 119-124 choline phosphotransferase 1 Homo sapiens 32-35 8070019-3 1994 The mechanisms of resistance to CPT-11 in PC-7/CPT-11 cells were reduced conversion of CPT-11 to its active metabolite SN-38 and point mutation topoisomerase I. Irinotecan 119-124 choline phosphotransferase 1 Homo sapiens 47-50 8070019-3 1994 The mechanisms of resistance to CPT-11 in PC-7/CPT-11 cells were reduced conversion of CPT-11 to its active metabolite SN-38 and point mutation topoisomerase I. Irinotecan 119-124 choline phosphotransferase 1 Homo sapiens 47-50 7505810-11 1994 With the use of rhG-CSF, the CPT-11 dose can be increased 33% above that in the original regimen (60 mg/m2 of CPT-11 and 80 mg/m2 of cisplatin). Irinotecan 29-35 colony stimulating factor 2 Homo sapiens 20-23 7505810-11 1994 With the use of rhG-CSF, the CPT-11 dose can be increased 33% above that in the original regimen (60 mg/m2 of CPT-11 and 80 mg/m2 of cisplatin). Irinotecan 110-116 colony stimulating factor 2 Homo sapiens 20-23 8476431-0 1993 Apoptosis of lung cancer cells caused by some anti-cancer agents (MMC, CPT-11, ADM) is inhibited by bcl-2. Irinotecan 71-77 BCL2 apoptosis regulator Homo sapiens 100-105 21584513-5 1992 Northern blot analysis showed about 5-fold increase in CAT mRNA levels in Kst-6 cells treated with CPT-11 or VP-16, but not with novobiocin. Irinotecan 99-105 catalase Homo sapiens 55-58 11578320-4 1993 In both cell lines, IFN-gamma at clinically achievable concentrations (10 and 100 U ml-1 enhanced the anti-proliferative activity of CPT-11 in the range of concentrations where CPT-11 showed more than 10% cell growth inhibition. Irinotecan 133-139 interferon gamma Homo sapiens 20-29 11578320-4 1993 In both cell lines, IFN-gamma at clinically achievable concentrations (10 and 100 U ml-1 enhanced the anti-proliferative activity of CPT-11 in the range of concentrations where CPT-11 showed more than 10% cell growth inhibition. Irinotecan 177-183 interferon gamma Homo sapiens 20-29 1332623-1 1992 CPT-11 and Topotecan are a new semisynthetic derivative of CPT, and have been shown to inhibit DNA topoisomerase I and to have a strong antitumor activity with low toxicity against murine tumor. Irinotecan 0-6 topoisomerase (DNA) I Mus musculus 95-114 21584513-3 1992 Exposure to inhibitors of DNA topoisomerase I (camptothecin: CPT-11) and II (etoposide: VP-16 and teniposide: VM-26) could efficiently induce CAT activities in both time- and dose-dependent manners. Irinotecan 61-67 host cell factor C1 Homo sapiens 88-93 21584513-6 1992 Proximal MDR1 promoter-binding activities of transacting factor were augmented in nuclear extracts from KB cells treated with CPT-11, VM-26, and VP-16. Irinotecan 126-132 ATP binding cassette subfamily B member 1 Homo sapiens 9-13 21584513-3 1992 Exposure to inhibitors of DNA topoisomerase I (camptothecin: CPT-11) and II (etoposide: VP-16 and teniposide: VM-26) could efficiently induce CAT activities in both time- and dose-dependent manners. Irinotecan 61-67 catalase Homo sapiens 142-145 1345810-7 1992 On the other hand, the conversion of CPT-11 to SN-38 in HAC2/0.1 was about 3-fold less than in HAC2/P. Irinotecan 37-43 hyperpolarization activated cyclic nucleotide gated potassium channel 1 Homo sapiens 56-64 1345810-7 1992 On the other hand, the conversion of CPT-11 to SN-38 in HAC2/0.1 was about 3-fold less than in HAC2/P. Irinotecan 37-43 hyperpolarization activated cyclic nucleotide gated potassium channel 1 Homo sapiens 56-60 1345810-9 1992 The accumulation of CPT-11 in HAC2/0.1 but not in HAC2/P was increased by BSO treatment. Irinotecan 20-26 hyperpolarization activated cyclic nucleotide gated potassium channel 1 Homo sapiens 30-38 1345810-7 1992 On the other hand, the conversion of CPT-11 to SN-38 in HAC2/0.1 was about 3-fold less than in HAC2/P. Irinotecan 47-52 hyperpolarization activated cyclic nucleotide gated potassium channel 1 Homo sapiens 56-64 1345810-9 1992 The accumulation of CPT-11 in HAC2/0.1 but not in HAC2/P was increased by BSO treatment. Irinotecan 20-26 hyperpolarization activated cyclic nucleotide gated potassium channel 1 Homo sapiens 30-34 1345810-10 1992 On the other hand, in HAC2/P the 50% inhibitory concentrations of SN-38 and CPT-11 were not influenced by BSO treatment. Irinotecan 66-71 hyperpolarization activated cyclic nucleotide gated potassium channel 1 Homo sapiens 22-26 1345810-10 1992 On the other hand, in HAC2/P the 50% inhibitory concentrations of SN-38 and CPT-11 were not influenced by BSO treatment. Irinotecan 76-82 hyperpolarization activated cyclic nucleotide gated potassium channel 1 Homo sapiens 22-26 1345810-11 1992 The 50% inhibitory concentration of CPT-11 for HAC2/0.1 was not reduced by BSO treatment to the level for HAC2/P, even though the GSH content had been reduced more than in HAC2/P. Irinotecan 36-42 hyperpolarization activated cyclic nucleotide gated potassium channel 1 Homo sapiens 47-55 1345810-11 1992 The 50% inhibitory concentration of CPT-11 for HAC2/0.1 was not reduced by BSO treatment to the level for HAC2/P, even though the GSH content had been reduced more than in HAC2/P. Irinotecan 36-42 hyperpolarization activated cyclic nucleotide gated potassium channel 1 Homo sapiens 47-51 1345810-7 1992 On the other hand, the conversion of CPT-11 to SN-38 in HAC2/0.1 was about 3-fold less than in HAC2/P. Irinotecan 47-52 hyperpolarization activated cyclic nucleotide gated potassium channel 1 Homo sapiens 56-60 1804390-0 1991 Augmentation of antiproliferative activity of CPT-11, a new derivative of camptothecin, by tumor necrosis factor against proliferation of gynecologic tumor cell lines. Irinotecan 46-52 tumor necrosis factor Homo sapiens 91-112 1309380-1 1992 PURPOSE: Camptothecin-11 (CPT-11) is a new semisynthetic derivative of CPT, and has been shown to inhibit DNA topoisomerase I and to have a strong antitumor activity with low toxicity in murine tumors. Irinotecan 9-24 topoisomerase (DNA) I Mus musculus 106-125 1309380-1 1992 PURPOSE: Camptothecin-11 (CPT-11) is a new semisynthetic derivative of CPT, and has been shown to inhibit DNA topoisomerase I and to have a strong antitumor activity with low toxicity in murine tumors. Irinotecan 26-32 topoisomerase (DNA) I Mus musculus 106-125 1804390-4 1991 In all four cell lines, rH-TNF at clinically achievable concentrations exhibited synergy with CPT-11. Irinotecan 94-100 tumor necrosis factor Homo sapiens 27-30 33805415-3 2021 We summarized meta-analyses, genome-wide association studies, clinical trials, drug labels, and guidelines relating to the impact of UGT1A1 polymorphisms on irinotecan, belinostat, pazopanib, or nilotinib toxicities. Irinotecan 157-167 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 133-139 33805415-4 2021 For irinotecan, UGT1A1*28 was significantly associated with neutropenia and diarrhea, particularly with doses >= 180 mg/m2, supporting the use of UGT1A1 to guide irinotecan prescribing. Irinotecan 162-172 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 16-22 33805415-4 2021 For irinotecan, UGT1A1*28 was significantly associated with neutropenia and diarrhea, particularly with doses >= 180 mg/m2, supporting the use of UGT1A1 to guide irinotecan prescribing. Irinotecan 162-172 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 146-152 33033581-11 2020 Knockdown of NeuroD1 enhanced the sensitivity to irinotecan of GNEC cell lines. Irinotecan 49-59 neuronal differentiation 1 Homo sapiens 13-20 33941341-0 2021 A four-component combination derived from Huang-Qin Decoction significantly enhances anticancer activity of irinotecan. Irinotecan 108-118 forkhead box G1 Homo sapiens 48-51 33941341-11 2021 In vitro assessment indicated that HB4 could enhance the effect of CPT-11 on inhibiting cell proliferation and inducing apoptosis in HCT116. Irinotecan 67-73 keratin 84 Homo sapiens 35-38 33941341-12 2021 Furthermore, the in vivo study confirmed that HB4 and HQD have similar pharmacological activity and could both enhance the antitumor effect of CPT-11 in HCT116 xenograft model. Irinotecan 143-149 keratin 84 Homo sapiens 46-49 33941341-13 2021 Meanwhile, HB4 could also reduce the CPT-11 induced GI toxicity. Irinotecan 37-43 keratin 84 Homo sapiens 11-14 33777142-13 2021 Conclusions: The oncological outcomes of patients with BRAF-mutated mCRC treated using FOLFIRI plus bevacizumab with irinotecan dose escalation as a first-line therapy are acceptable with tolerable AEs; this may be a feasible treatment option in such patients. Irinotecan 117-127 B-Raf proto-oncogene, serine/threonine kinase Homo sapiens 55-59 33235468-0 2020 Tumor Response to Irinotecan is Associated with IL-10 Expression Level in Metastatic Colorectal Cancer-Results from mCRC Biomarker Study. Irinotecan 18-28 interleukin 10 Homo sapiens 48-53 33235468-11 2020 By utilizing univariate and multivariate Cox proportional hazard analyses, we found that low IL-10 level in plasma was significantly associated with improved overall survival (OS) of mCRC patients treated with irinotecan-containing regimen-with optimal cutoff value of 5.525pg/mL, respectively (p =0.002). Irinotecan 210-220 interleukin 10 Homo sapiens 93-98 33235468-12 2020 In addition, the low IL-10 expression level in tumor tissue was significantly associated with the improved OS for the irinotecan-containing regimen (p = 0.023). Irinotecan 118-128 interleukin 10 Homo sapiens 21-26 33235468-13 2020 Conclusion: Our study demonstrated that IL-10 could act as a prognostic biomarker for mCRC patients undergoing irinotecan-containing chemotherapy. Irinotecan 111-121 interleukin 10 Homo sapiens 40-45 33235483-7 2020 Prevalent UGT1A1 gene [TA]7TAA promoter allelic variant UGT1A1*28, characterized by an extra TA repeat, is associated with low transcriptional and reduced enzymatic effectiveness, decreased SN38 active irinotecan metabolite glucuronidation, vs wild-type UGT1A1*1 [A(TA)6TAA]. Irinotecan 202-212 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 10-16 33235483-7 2020 Prevalent UGT1A1 gene [TA]7TAA promoter allelic variant UGT1A1*28, characterized by an extra TA repeat, is associated with low transcriptional and reduced enzymatic effectiveness, decreased SN38 active irinotecan metabolite glucuronidation, vs wild-type UGT1A1*1 [A(TA)6TAA]. Irinotecan 202-212 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 56-62 33235483-7 2020 Prevalent UGT1A1 gene [TA]7TAA promoter allelic variant UGT1A1*28, characterized by an extra TA repeat, is associated with low transcriptional and reduced enzymatic effectiveness, decreased SN38 active irinotecan metabolite glucuronidation, vs wild-type UGT1A1*1 [A(TA)6TAA]. Irinotecan 202-212 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 56-62 33235483-11 2020 Implementation of up-front evaluation of the five validated DPYD variants and UGT1A1*28 in the multidisciplinary molecular tumor board, also including CRC genetic characterization, addresses potential treatments with fluoropyrimidines and irinotecan associations at proper doses and schedules, particularly for early CRC, MCRC patients fit for intensive regimens or unfit for conventional regimens, requiring treatment modulations, and also for patients who experience severe, unexpected toxicities. Irinotecan 239-249 dihydropyrimidine dehydrogenase Homo sapiens 60-64 33235483-11 2020 Implementation of up-front evaluation of the five validated DPYD variants and UGT1A1*28 in the multidisciplinary molecular tumor board, also including CRC genetic characterization, addresses potential treatments with fluoropyrimidines and irinotecan associations at proper doses and schedules, particularly for early CRC, MCRC patients fit for intensive regimens or unfit for conventional regimens, requiring treatment modulations, and also for patients who experience severe, unexpected toxicities. Irinotecan 239-249 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 78-84 33777142-0 2021 UGT1A1 Polymorphism for Irinotecan Dose Escalation in Patients with BRAF-Mutated Metastatic Colorectal Cancer Treated with First-Line Bevacizumab and FOLFIRI. Irinotecan 24-34 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 33777142-0 2021 UGT1A1 Polymorphism for Irinotecan Dose Escalation in Patients with BRAF-Mutated Metastatic Colorectal Cancer Treated with First-Line Bevacizumab and FOLFIRI. Irinotecan 24-34 B-Raf proto-oncogene, serine/threonine kinase Homo sapiens 68-72 33777142-3 2021 In this study, we explored the effects and oncological outcomes of UGT1A1 polymorphism for irinotecan escalation in patients with BRAF-mutated mCRC. Irinotecan 91-101 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 67-73 33777142-3 2021 In this study, we explored the effects and oncological outcomes of UGT1A1 polymorphism for irinotecan escalation in patients with BRAF-mutated mCRC. Irinotecan 91-101 B-Raf proto-oncogene, serine/threonine kinase Homo sapiens 130-134 33793796-0 2021 Possible roles of intestinal P-glycoprotein and cytochrome P450 3A on the limited oral absorption of irinotecan. Irinotecan 101-111 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 29-43 33793796-0 2021 Possible roles of intestinal P-glycoprotein and cytochrome P450 3A on the limited oral absorption of irinotecan. Irinotecan 101-111 cytochrome P450, family 3, subfamily a, polypeptide 62 Rattus norvegicus 48-66 33793796-6 2021 The pharmacokinetics of irinotecan was investigated when verapamil, an inhibitor of the P-glycoprotein (P-gp) and cytochrome P450 3A (CYP3A) was pre-administered. Irinotecan 24-34 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 88-102 33793796-6 2021 The pharmacokinetics of irinotecan was investigated when verapamil, an inhibitor of the P-glycoprotein (P-gp) and cytochrome P450 3A (CYP3A) was pre-administered. Irinotecan 24-34 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 104-108 33793796-6 2021 The pharmacokinetics of irinotecan was investigated when verapamil, an inhibitor of the P-glycoprotein (P-gp) and cytochrome P450 3A (CYP3A) was pre-administered. Irinotecan 24-34 cytochrome P450, family 3, subfamily a, polypeptide 62 Rattus norvegicus 114-132 33793796-6 2021 The pharmacokinetics of irinotecan was investigated when verapamil, an inhibitor of the P-glycoprotein (P-gp) and cytochrome P450 3A (CYP3A) was pre-administered. Irinotecan 24-34 cytochrome P450, family 3, subfamily a, polypeptide 62 Rattus norvegicus 134-139 33793796-7 2021 KEY FINDINGS: The intestinal absorption of irinotecan was enhanced in the presence of verapamil, indicating that efflux by intestinal P-gp contributes to its limited oral absorption. Irinotecan 43-53 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 134-138 33793796-10 2021 CONCLUSION: The findings presented herein suggest that intestinal efflux by P-gp is mainly and intestinal metabolism by CYP3A is partially responsible for the limited oral absorption of irinotecan. Irinotecan 186-196 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 76-80 33793796-10 2021 CONCLUSION: The findings presented herein suggest that intestinal efflux by P-gp is mainly and intestinal metabolism by CYP3A is partially responsible for the limited oral absorption of irinotecan. Irinotecan 186-196 cytochrome P450, family 3, subfamily a, polypeptide 62 Rattus norvegicus 120-125 33790151-0 2021 [The Efficacy of topo I-pS10 Expression in Gastric Cancer as a Predictive Biomarker for Irinotecan Use]. Irinotecan 88-98 taste 2 receptor member 12 pseudogene Homo sapiens 24-28 33790151-3 2021 We have developed an immunohistochemical staining-based biomarker; topo I-pS10, for predicting irinotecan efficacy. Irinotecan 95-105 taste 2 receptor member 12 pseudogene Homo sapiens 74-78 33790151-13 2021 CONCLUSION: topo I-pS10 staining can be used as a predictive biomarker for irinotecan for gastric cancer patients. Irinotecan 75-85 taste 2 receptor member 12 pseudogene Homo sapiens 19-23 34718178-6 2022 Impressively, BI@PEG-SN38 nanoparticles effectively reverse chemoresistance to CPT-11 (resistance index dropping from ~274.00-456.00 to ~1.70-4.68) and PEG-S-S-SN38 (resistance index dropping from ~5.83-14.00 to ~1.70-4.68) in three BCRP-overexpressing cancer cell lines. Irinotecan 79-85 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 233-237 26386386-3 2015 Overexpression of a multidrug resistance (MDR) transporter ABCG2 in vitro has been shown to cause resistance to 5-fluorouracil (5-FU) and irinotecan, components of the most commonly adopted regimens for treating CRC. Irinotecan 138-148 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 59-64 34854953-15 2022 CONCLUSION: Tlr4DeltaIEC and WT had no baseline compositional microbiome differences, but functional differences at baseline and following irinotecan. Irinotecan 139-149 toll-like receptor 4 Mus musculus 12-24 34699766-0 2022 The PPARgamma-dependent effect of flavonoid luteolin against damage induced by the chemotherapeutic irinotecan in human intestinal cells. Irinotecan 100-110 peroxisome proliferator activated receptor gamma Homo sapiens 4-13 34699766-5 2022 Irinotecan downregulated PPARgamma expression and upregulated inflammatory and oxidative genes, while luteolin upregulated PPARgamma, HO-1, SOD and decreased expression of IL-1beta and iNOS. Irinotecan 0-10 peroxisome proliferator activated receptor gamma Homo sapiens 25-34 34699766-8 2022 The present study showed that the protective effect of luteolin against irinotecan is PPARgamma dependent. Irinotecan 72-82 peroxisome proliferator activated receptor gamma Homo sapiens 86-95 34968866-0 2022 Phospho-Aspirin (MDC-22) inhibits pancreatic cancer growth in patient-derived tumor xenografts and KPC mice by targeting EGFR: Enhanced efficacy in combination with irinotecan. Irinotecan 165-175 C-C motif chemokine ligand 22 Homo sapiens 17-20 34968866-8 2022 In human pancreatic cancer cell lines, MDC-22 enhanced the growth inhibitory effect of irinotecan, and to a lesser degree those of gemcitabine and nab-paclitaxel. Irinotecan 87-97 C-C motif chemokine ligand 22 Homo sapiens 39-42 34968866-10 2022 Furthermore, MDC-22 enhanced irinotecan"s effect on cell migration, in part, by inhibiting EGFR/FAK signaling. Irinotecan 29-39 epidermal growth factor receptor Homo sapiens 91-95 34968866-10 2022 Furthermore, MDC-22 enhanced irinotecan"s effect on cell migration, in part, by inhibiting EGFR/FAK signaling. Irinotecan 29-39 protein tyrosine kinase 2 Homo sapiens 96-99 34718178-7 2022 More importantly, reversal of BCRP-mediated chemoresistance to CPT-11 (P values lower than 0.001-0.0001) and PEG-S-S-SN38 (P values lower than 0.01-0.001) by BI@PEG-SN38 nanoparticles are further confirmed with two panels of colorectal cancer xenograft models in vivo. Irinotecan 63-69 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 30-34 34718178-10 2022 By generating panels of colorectal cancer models, we demonstrate that BI@PEG-SN38 nanoparticles effectively and selectively reversed BCRP-mediated tumor resistance to SN38/CPT-11 in vitro and in vivo. Irinotecan 172-178 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 133-137 34689170-10 2022 CONCLUSIONS: TIMELESS rs2291739 might have different effects on therapeutic efficacy between oxaliplatin- and irinotecan-based chemotherapies. Irinotecan 110-120 timeless circadian regulator Homo sapiens 13-21 34700030-3 2022 METHODS: We performed next generation sequencing to study the evolution of KRAS wild-type CRC organoids during adaptation to irinotecan-based chemotherapy combined with EGFR-inhibition. Irinotecan 125-135 KRAS proto-oncogene, GTPase Homo sapiens 75-79 34667024-9 2022 SN-38 had the lowest IC50 (approximately 10 nM), and its pro-apoptotic effects were countered by knockdown of CEBPA but not of TP53 Irinotecan significantly inhibited tumor growth at well tolerated doses, induced nuclear expression of C/EBPalpha, and inhibited HIF1a expression in DDLS patient-derived and cancer cell line xenograft models. Irinotecan 0-5 CCAAT enhancer binding protein alpha Homo sapiens 110-115 34667024-9 2022 SN-38 had the lowest IC50 (approximately 10 nM), and its pro-apoptotic effects were countered by knockdown of CEBPA but not of TP53 Irinotecan significantly inhibited tumor growth at well tolerated doses, induced nuclear expression of C/EBPalpha, and inhibited HIF1a expression in DDLS patient-derived and cancer cell line xenograft models. Irinotecan 0-5 CCAAT enhancer binding protein alpha Homo sapiens 235-245 34667024-9 2022 SN-38 had the lowest IC50 (approximately 10 nM), and its pro-apoptotic effects were countered by knockdown of CEBPA but not of TP53 Irinotecan significantly inhibited tumor growth at well tolerated doses, induced nuclear expression of C/EBPalpha, and inhibited HIF1a expression in DDLS patient-derived and cancer cell line xenograft models. Irinotecan 0-5 hypoxia inducible factor 1 subunit alpha Homo sapiens 261-266 34667024-9 2022 SN-38 had the lowest IC50 (approximately 10 nM), and its pro-apoptotic effects were countered by knockdown of CEBPA but not of TP53 Irinotecan significantly inhibited tumor growth at well tolerated doses, induced nuclear expression of C/EBPalpha, and inhibited HIF1a expression in DDLS patient-derived and cancer cell line xenograft models. Irinotecan 132-142 CCAAT enhancer binding protein alpha Homo sapiens 235-245 34667024-9 2022 SN-38 had the lowest IC50 (approximately 10 nM), and its pro-apoptotic effects were countered by knockdown of CEBPA but not of TP53 Irinotecan significantly inhibited tumor growth at well tolerated doses, induced nuclear expression of C/EBPalpha, and inhibited HIF1a expression in DDLS patient-derived and cancer cell line xenograft models. Irinotecan 132-142 hypoxia inducible factor 1 subunit alpha Homo sapiens 261-266 34667024-12 2022 These include irinotecan, which induces apoptosis of DDLS cells in a C/EBPalpha-dependent, p53-independent manner and should be clinically evaluated in patients with advanced DDLS. Irinotecan 14-24 CCAAT enhancer binding protein alpha Homo sapiens 69-79 34667024-12 2022 These include irinotecan, which induces apoptosis of DDLS cells in a C/EBPalpha-dependent, p53-independent manner and should be clinically evaluated in patients with advanced DDLS. Irinotecan 14-24 tumor protein p53 Homo sapiens 91-94 34860573-0 2022 All You Need to Know About UGT1A1 Genetic Testing for Patients Treated With Irinotecan: A Practitioner-Friendly Guide. Irinotecan 76-86 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 27-33 34697081-12 2022 The use of tissue-specific humanized models expressing UGT1A1 in liver or intestine has confirmed the relevance of the intestinal tract in the detoxification of irinotecan. Irinotecan 161-171 UDP glucuronosyltransferase 1 family, polypeptide A1 Mus musculus 55-61 34911343-18 2022 NAT2 590G>A appears to be a potential prognostic factor for OS of SCLC patients after irinotecan therapy and 481C>T came out to be significant factor using CART. Irinotecan 86-96 N-acetyltransferase 2 Homo sapiens 0-4 34941047-1 2021 BACKGROUND: Combined treatment with anlotinib, irinotecan, as well as vincristine for advanced Ewing sarcoma (EWS) has been verified been effective in the prospective trial of Peking University People"s Hospital EWS trial-02. Irinotecan 47-57 EWS RNA binding protein 1 Homo sapiens 212-215 34941047-11 2021 CONCLUSION: QoL exhibited a trend towards improvement in accordance with high objective response in this trial with the receipt of combination therapy of anlotinib, vinsristine, and irinotecan for advanced EWS. Irinotecan 182-192 EWS RNA binding protein 1 Homo sapiens 206-209 34697081-5 2022 Mice were challenged with irinotecan (CPT-11), a prodrug hydrolyzed by carboxylesterases to form the active metabolite SN-38 and detoxified by UGT1A1. Irinotecan 26-36 UDP glucuronosyltransferase 1 family, polypeptide A1 Mus musculus 143-149 34697081-5 2022 Mice were challenged with irinotecan (CPT-11), a prodrug hydrolyzed by carboxylesterases to form the active metabolite SN-38 and detoxified by UGT1A1. Irinotecan 38-44 UDP glucuronosyltransferase 1 family, polypeptide A1 Mus musculus 143-149 34697081-7 2022 When exposed to a low dose of CPT-11 (10 mg/kg), hUGT1A1HEP mice displayed greater intestinal inflammatory (IL-1beta and IL-6) insult in addition to p53-triggered apoptotic responses. Irinotecan 30-36 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 49-56 34697081-7 2022 When exposed to a low dose of CPT-11 (10 mg/kg), hUGT1A1HEP mice displayed greater intestinal inflammatory (IL-1beta and IL-6) insult in addition to p53-triggered apoptotic responses. Irinotecan 30-36 interleukin 1 alpha Mus musculus 108-116 34697081-7 2022 When exposed to a low dose of CPT-11 (10 mg/kg), hUGT1A1HEP mice displayed greater intestinal inflammatory (IL-1beta and IL-6) insult in addition to p53-triggered apoptotic responses. Irinotecan 30-36 interleukin 6 Mus musculus 121-125 34697081-7 2022 When exposed to a low dose of CPT-11 (10 mg/kg), hUGT1A1HEP mice displayed greater intestinal inflammatory (IL-1beta and IL-6) insult in addition to p53-triggered apoptotic responses. Irinotecan 30-36 transformation related protein 53, pseudogene Mus musculus 149-152 34725191-0 2022 Combination therapy of hepatocellular carcinoma by GPC3-targeted bispecific antibody and Irinotecan is potent in suppressing tumor growth in mice. Irinotecan 89-99 glypican 3 Mus musculus 51-55 34971802-1 2022 OBJECTIVE: Intra-tumoral expression of the serine hydrolase carboxylesterase 2 (CES2) contributes to the activation of the pro-drug irinotecan in pancreatic ductal adenocarcinoma (PDAC). Irinotecan 132-142 carboxylesterase 2H Mus musculus 60-78 34971802-1 2022 OBJECTIVE: Intra-tumoral expression of the serine hydrolase carboxylesterase 2 (CES2) contributes to the activation of the pro-drug irinotecan in pancreatic ductal adenocarcinoma (PDAC). Irinotecan 132-142 carboxylesterase 2H Mus musculus 80-84 34860573-2 2022 Severe neutropenia and diarrhea are common dose-limiting toxicities of irinotecan-based therapy, and UGT1A1 polymorphisms are one of the major risk factors of these toxicities. Irinotecan 71-81 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 101-107 34860573-3 2022 In 2005, the US Food and Drug Administration revised the drug label to indicate that patients with UGT1A1*28 homozygous genotype should receive a decreased dose of irinotecan. Irinotecan 164-174 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 99-105 34811961-10 2021 Alkaline phosphatase (ALP), SA, and WBC counts were important predictors of survival among patients treated with second-line liposomal irinotecan. Irinotecan 135-145 alkaline phosphatase, placental Homo sapiens 0-20 34811961-10 2021 Alkaline phosphatase (ALP), SA, and WBC counts were important predictors of survival among patients treated with second-line liposomal irinotecan. Irinotecan 135-145 alkaline phosphatase, placental Homo sapiens 22-25 34894942-3 2021 Herein, we reported a novel strategy to construct highly releasable and structurally stable SN-38-conjugates, in which CTSB linkers directly connected to the 10-OH group through ether bond, not to the common 20-OH group of lactones of SN-38. Irinotecan 92-97 cathepsin B Homo sapiens 119-123 34651524-3 2021 Sacituzumab govitecan (IMMU-132) is an antibody drug conjugate (ADC) comprised of an active metabolite of irinotecan, SN-38, bound to a humanized monoclonal antibody which targets trophoblastic cell-surface antigen 2 (Trop-2). Irinotecan 106-116 tumor associated calcium signal transducer 2 Homo sapiens 218-224 34075792-0 2021 Silencing of E-cadherin expression leads to increased chemosensitivity to irinotecan and oxaliplatin in colorectal cancer cell lines. Irinotecan 74-84 cadherin 1 Homo sapiens 13-23 34651524-3 2021 Sacituzumab govitecan (IMMU-132) is an antibody drug conjugate (ADC) comprised of an active metabolite of irinotecan, SN-38, bound to a humanized monoclonal antibody which targets trophoblastic cell-surface antigen 2 (Trop-2). Irinotecan 118-123 tumor associated calcium signal transducer 2 Homo sapiens 218-224 34887262-7 2021 We determined the efficacy of low-dose SN-38 or metformin in sensitizing unresponsive tumors to respond to anti-PD-1 therapy in a syngeneic tumor system. Irinotecan 39-44 programmed cell death 1 Homo sapiens 112-116 34258881-0 2021 Mechanistic insights into p53-regulated cytotoxicity of combined entinostat and irinotecan against colorectal cancer cells. Irinotecan 80-90 tumor protein p53 Homo sapiens 26-29 34887262-8 2021 We deciphered novel therapeutic mechanisms underlying the SN-38 and anti-PD-1 therapy-mediated engagement of natural killer (NK) or CD8+ T cells to infiltrate tumors and boost antitumor immunity. Irinotecan 58-63 CD8a molecule Homo sapiens 132-135 34887262-10 2021 Low-dose SN-38 or metformin abrogated PD-L1 protein expression, promoted FOXO3 protein level, and significantly increased the animal survival rate in syngeneic mouse tumor models. Irinotecan 9-14 CD274 antigen Mus musculus 38-43 34887262-10 2021 Low-dose SN-38 or metformin abrogated PD-L1 protein expression, promoted FOXO3 protein level, and significantly increased the animal survival rate in syngeneic mouse tumor models. Irinotecan 9-14 forkhead box O3 Mus musculus 73-78 34887262-11 2021 SN-38 or metformin sensitized unresponsive tumors responding to anti-PD-1 therapy by engaging NK or CD8+ T cells to infiltrate the tumor microenvironment (TME) and secret interferon-gamma and granzyme B to kill tumors. Irinotecan 0-5 programmed cell death 1 Mus musculus 69-73 34887262-11 2021 SN-38 or metformin sensitized unresponsive tumors responding to anti-PD-1 therapy by engaging NK or CD8+ T cells to infiltrate the tumor microenvironment (TME) and secret interferon-gamma and granzyme B to kill tumors. Irinotecan 0-5 CD8a molecule Homo sapiens 100-103 34887262-11 2021 SN-38 or metformin sensitized unresponsive tumors responding to anti-PD-1 therapy by engaging NK or CD8+ T cells to infiltrate the tumor microenvironment (TME) and secret interferon-gamma and granzyme B to kill tumors. Irinotecan 0-5 interferon gamma Homo sapiens 171-187 34887262-11 2021 SN-38 or metformin sensitized unresponsive tumors responding to anti-PD-1 therapy by engaging NK or CD8+ T cells to infiltrate the tumor microenvironment (TME) and secret interferon-gamma and granzyme B to kill tumors. Irinotecan 0-5 granzyme B Homo sapiens 192-202 34887262-12 2021 SN-38 suppressed the levels of c-Myc and STAT3 proteins, which controlled PD-L1 expression. Irinotecan 0-5 MYC proto-oncogene, bHLH transcription factor Homo sapiens 31-36 34887262-12 2021 SN-38 suppressed the levels of c-Myc and STAT3 proteins, which controlled PD-L1 expression. Irinotecan 0-5 signal transducer and activator of transcription 3 Mus musculus 41-46 34887262-12 2021 SN-38 suppressed the levels of c-Myc and STAT3 proteins, which controlled PD-L1 expression. Irinotecan 0-5 CD274 antigen Mus musculus 74-79 34887262-15 2021 CONCLUSION: We show that SN-38 or metformin can boost antitumor immunity in the TME by inhibiting c-Myc and STAT3 through FOXO3 activation. Irinotecan 25-30 MYC proto-oncogene, bHLH transcription factor Homo sapiens 98-103 34887262-15 2021 CONCLUSION: We show that SN-38 or metformin can boost antitumor immunity in the TME by inhibiting c-Myc and STAT3 through FOXO3 activation. Irinotecan 25-30 signal transducer and activator of transcription 3 Homo sapiens 108-113 34887262-15 2021 CONCLUSION: We show that SN-38 or metformin can boost antitumor immunity in the TME by inhibiting c-Myc and STAT3 through FOXO3 activation. Irinotecan 25-30 forkhead box O3 Homo sapiens 122-127 34258881-5 2021 The topoisomerase-1 inhibitor irinotecan induces acetylation of several lysine residues within p53. Irinotecan 30-40 transformation related protein 53, pseudogene Mus musculus 95-98 34258881-6 2021 Inhibition of histone deacetylaces (HDACs) with the class I HDAC inhibitor entinostat synergistically triggers mitochondrial damage and apoptosis in irinotecan-treated p53-positive CRC cells. Irinotecan 149-159 transformation related protein 53, pseudogene Mus musculus 168-171 34847314-4 2021 Additional, well-planned trials of the UGT1A1 genotype-based approach to irinotecan therapy are predicted to reduce adverse drug events in people with the UGT1A1*28/*28 genotypes and improve treatment efficacy in the rest of the patients, which might be cost-effective. Irinotecan 73-83 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 39-45 34676875-0 2021 Knockdown of TRIM9 attenuates irinotecan-induced intestinal mucositis in IEC-6 cells by regulating DUSP6 expression via the P38 pathway. Irinotecan 30-40 tripartite motif-containing 9 Rattus norvegicus 13-18 34676875-0 2021 Knockdown of TRIM9 attenuates irinotecan-induced intestinal mucositis in IEC-6 cells by regulating DUSP6 expression via the P38 pathway. Irinotecan 30-40 dual specificity phosphatase 6 Rattus norvegicus 99-104 34676875-0 2021 Knockdown of TRIM9 attenuates irinotecan-induced intestinal mucositis in IEC-6 cells by regulating DUSP6 expression via the P38 pathway. Irinotecan 30-40 mitogen activated protein kinase 14 Rattus norvegicus 124-127 34676875-3 2021 The present study aimed to investigate the effect of TRIM9 on irinotecan-induced intestinal mucositis in the rat intestinal epithelial cell line IEC-6. Irinotecan 62-72 tripartite motif-containing 9 Rattus norvegicus 53-58 34676875-6 2021 Treatment with irinotecan significantly inhibited cell proliferation and induced cell apoptosis, TRIM9 expression, intestinal mucosal barrier impairment, the levels of inflammatory cytokines and P38 phosphorylation in IEC-6 cells, while the expression levels of epithelial barrier tight-junction protein ZO-1 and Claudin-4 were decreased. Irinotecan 15-25 tripartite motif-containing 9 Rattus norvegicus 97-102 34676875-6 2021 Treatment with irinotecan significantly inhibited cell proliferation and induced cell apoptosis, TRIM9 expression, intestinal mucosal barrier impairment, the levels of inflammatory cytokines and P38 phosphorylation in IEC-6 cells, while the expression levels of epithelial barrier tight-junction protein ZO-1 and Claudin-4 were decreased. Irinotecan 15-25 mitogen activated protein kinase 14 Rattus norvegicus 195-198 34676875-6 2021 Treatment with irinotecan significantly inhibited cell proliferation and induced cell apoptosis, TRIM9 expression, intestinal mucosal barrier impairment, the levels of inflammatory cytokines and P38 phosphorylation in IEC-6 cells, while the expression levels of epithelial barrier tight-junction protein ZO-1 and Claudin-4 were decreased. Irinotecan 15-25 tight junction protein 1 Rattus norvegicus 304-308 34676875-6 2021 Treatment with irinotecan significantly inhibited cell proliferation and induced cell apoptosis, TRIM9 expression, intestinal mucosal barrier impairment, the levels of inflammatory cytokines and P38 phosphorylation in IEC-6 cells, while the expression levels of epithelial barrier tight-junction protein ZO-1 and Claudin-4 were decreased. Irinotecan 15-25 claudin 4 Rattus norvegicus 313-322 34676875-7 2021 Knockdown of TRIM9 partly counteracted the effect of irinotecan treatment, and inhibition of P38 potently reversed the effect of TRIM9 overexpression in IEC-6 cells. Irinotecan 53-63 tripartite motif-containing 9 Rattus norvegicus 13-18 34847314-4 2021 Additional, well-planned trials of the UGT1A1 genotype-based approach to irinotecan therapy are predicted to reduce adverse drug events in people with the UGT1A1*28/*28 genotypes and improve treatment efficacy in the rest of the patients, which might be cost-effective. Irinotecan 73-83 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 155-161 34917992-7 2021 Conclusions: EGFR tyrosine inhibitors plus irinotecan plus cisplatin chemotherapy might be a potential treatment option for advanced pulmonary neuroendocrine neoplasms harboring EGFR mutations. Irinotecan 43-53 epidermal growth factor receptor Homo sapiens 178-182 34815254-8 2022 The small molecule exportin-1 inhibitor selinexor in combination with cisplatin or irinotecan dramatically inhibited tumor growth in chemonaive and chemorelapsed SCLC patient-derived xenografts, respectively. Irinotecan 83-93 exportin 1 Homo sapiens 19-29 34794220-1 2021 Objective: To investigate the correlation between UGT1A1 polymorphisms and the irinotecan plus S-1 regimen-induced toxicities in Chinese advanced esophageal squamous cell carcinoma (ESCC) patients. Irinotecan 79-89 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 50-56 34794220-5 2021 Irinotecan plus S-1 regimen-induced toxicities of patients with different UGT1A1 polymorphisms were observed. Irinotecan 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 74-80 34830383-8 2021 We found that the phenylfurocoumarin derivative (R)-9-(3,4-dimethoxyphenyl)-4-((3,3-dimethyloxiran-2-yl)methoxy)-7H-furo (3,2-g)chromen-7-one (PFC) significantly decreased the IC50 of SN-38 in HCT-116/BCRP colon cancer cells. Irinotecan 184-189 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 201-205 34830244-9 2021 p130Cas was inducible by 5-fluorouracil (5-FU) and FOLFIRI (folinic acid, 5-FU, irinotecan), and p130Cas and EREG were upregulated in distant metastases (GSE121418). Irinotecan 80-90 BCAR1 scaffold protein, Cas family member Homo sapiens 0-7 34795594-0 2021 Berberine Improves Irinotecan-Induced Intestinal Mucositis Without Impairing the Anti-colorectal Cancer Efficacy of Irinotecan by Inhibiting Bacterial beta-glucuronidase. Irinotecan 116-126 glucuronidase beta Homo sapiens 151-169 34474344-0 2021 LEF1 silencing sensitizes colorectal cancer cells to oxaliplatin, 5-FU, and irinotecan. Irinotecan 76-86 lymphoid enhancer binding factor 1 Homo sapiens 0-4 34327766-0 2021 Impact of UGT1A1 Genotype on the Efficacy and Safety of Irinotecan-based Chemotherapy in Metastatic Colorectal Cancer. Irinotecan 56-66 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 10-16 34327766-2 2021 We evaluated the associations between the UGT1A1 genotype linked to adverse events - caused by irinotecan - and the efficacy and safety of mXELIRI and FOLFIRI. Irinotecan 95-105 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 42-48 34536357-6 2021 CES2 mRNA level was significantly increased by double knockdown of METTL3 and METTL14 but was decreased by knockdown of FTO or ALKBH5 in HepaRG and HepG2 cells, leading to changes in its protein level and hydrolase activity for 7-ethyl-10-(4-(1-piperidino)-1-piperidino)carbonyloxycamptothecin (CPT-11), suggesting that m6A modification negatively regulates CES2 expression. Irinotecan 228-293 carboxylesterase 2 Homo sapiens 0-4 34536357-6 2021 CES2 mRNA level was significantly increased by double knockdown of METTL3 and METTL14 but was decreased by knockdown of FTO or ALKBH5 in HepaRG and HepG2 cells, leading to changes in its protein level and hydrolase activity for 7-ethyl-10-(4-(1-piperidino)-1-piperidino)carbonyloxycamptothecin (CPT-11), suggesting that m6A modification negatively regulates CES2 expression. Irinotecan 228-293 methyltransferase 3, N6-adenosine-methyltransferase complex catalytic subunit Homo sapiens 67-73 34536357-6 2021 CES2 mRNA level was significantly increased by double knockdown of METTL3 and METTL14 but was decreased by knockdown of FTO or ALKBH5 in HepaRG and HepG2 cells, leading to changes in its protein level and hydrolase activity for 7-ethyl-10-(4-(1-piperidino)-1-piperidino)carbonyloxycamptothecin (CPT-11), suggesting that m6A modification negatively regulates CES2 expression. Irinotecan 228-293 methyltransferase 14, N6-adenosine-methyltransferase subunit Homo sapiens 78-85 34536357-6 2021 CES2 mRNA level was significantly increased by double knockdown of METTL3 and METTL14 but was decreased by knockdown of FTO or ALKBH5 in HepaRG and HepG2 cells, leading to changes in its protein level and hydrolase activity for 7-ethyl-10-(4-(1-piperidino)-1-piperidino)carbonyloxycamptothecin (CPT-11), suggesting that m6A modification negatively regulates CES2 expression. Irinotecan 228-293 FTO alpha-ketoglutarate dependent dioxygenase Homo sapiens 120-123 34536357-6 2021 CES2 mRNA level was significantly increased by double knockdown of METTL3 and METTL14 but was decreased by knockdown of FTO or ALKBH5 in HepaRG and HepG2 cells, leading to changes in its protein level and hydrolase activity for 7-ethyl-10-(4-(1-piperidino)-1-piperidino)carbonyloxycamptothecin (CPT-11), suggesting that m6A modification negatively regulates CES2 expression. Irinotecan 228-293 alkB homolog 5, RNA demethylase Homo sapiens 127-133 34536357-6 2021 CES2 mRNA level was significantly increased by double knockdown of METTL3 and METTL14 but was decreased by knockdown of FTO or ALKBH5 in HepaRG and HepG2 cells, leading to changes in its protein level and hydrolase activity for 7-ethyl-10-(4-(1-piperidino)-1-piperidino)carbonyloxycamptothecin (CPT-11), suggesting that m6A modification negatively regulates CES2 expression. Irinotecan 295-301 carboxylesterase 2 Homo sapiens 0-4 34536357-6 2021 CES2 mRNA level was significantly increased by double knockdown of METTL3 and METTL14 but was decreased by knockdown of FTO or ALKBH5 in HepaRG and HepG2 cells, leading to changes in its protein level and hydrolase activity for 7-ethyl-10-(4-(1-piperidino)-1-piperidino)carbonyloxycamptothecin (CPT-11), suggesting that m6A modification negatively regulates CES2 expression. Irinotecan 295-301 methyltransferase 3, N6-adenosine-methyltransferase complex catalytic subunit Homo sapiens 67-73 34536357-6 2021 CES2 mRNA level was significantly increased by double knockdown of METTL3 and METTL14 but was decreased by knockdown of FTO or ALKBH5 in HepaRG and HepG2 cells, leading to changes in its protein level and hydrolase activity for 7-ethyl-10-(4-(1-piperidino)-1-piperidino)carbonyloxycamptothecin (CPT-11), suggesting that m6A modification negatively regulates CES2 expression. Irinotecan 295-301 methyltransferase 14, N6-adenosine-methyltransferase subunit Homo sapiens 78-85 34536357-6 2021 CES2 mRNA level was significantly increased by double knockdown of METTL3 and METTL14 but was decreased by knockdown of FTO or ALKBH5 in HepaRG and HepG2 cells, leading to changes in its protein level and hydrolase activity for 7-ethyl-10-(4-(1-piperidino)-1-piperidino)carbonyloxycamptothecin (CPT-11), suggesting that m6A modification negatively regulates CES2 expression. Irinotecan 295-301 FTO alpha-ketoglutarate dependent dioxygenase Homo sapiens 120-123 34536357-6 2021 CES2 mRNA level was significantly increased by double knockdown of METTL3 and METTL14 but was decreased by knockdown of FTO or ALKBH5 in HepaRG and HepG2 cells, leading to changes in its protein level and hydrolase activity for 7-ethyl-10-(4-(1-piperidino)-1-piperidino)carbonyloxycamptothecin (CPT-11), suggesting that m6A modification negatively regulates CES2 expression. Irinotecan 295-301 alkB homolog 5, RNA demethylase Homo sapiens 127-133 34536357-6 2021 CES2 mRNA level was significantly increased by double knockdown of METTL3 and METTL14 but was decreased by knockdown of FTO or ALKBH5 in HepaRG and HepG2 cells, leading to changes in its protein level and hydrolase activity for 7-ethyl-10-(4-(1-piperidino)-1-piperidino)carbonyloxycamptothecin (CPT-11), suggesting that m6A modification negatively regulates CES2 expression. Irinotecan 295-301 carboxylesterase 2 Homo sapiens 358-362 34474344-10 2021 In summary, we showed the role of LEF1 in chemo-resistance of colorectal cancer cells to Oxaliplatin, Irinotecan and 5-FU. Irinotecan 102-112 lymphoid enhancer binding factor 1 Homo sapiens 34-38 34703007-5 2022 RESULTS: Irinotecan clearance was influenced by rs4149057 in SLCO1B1, body surface area, and co-administration of 5-fluorouracil/leucovorin/bevacizumab. Irinotecan 9-19 solute carrier organic anion transporter family member 1B1 Homo sapiens 61-68 34365218-5 2021 Further, majority of the non-platinum drugs except irinotecan increased ERK1/2 activation in platinum-taxol resistant cells as observed by live-cell BRET assessment which were associated with p90RSK1/2 and BAD activation along with upregulation of multidrug transporter gene ABCC1 and cell survival genes like cyclin D1 and Bcl2. Irinotecan 51-61 mitogen-activated protein kinase 3 Homo sapiens 72-78 34365218-5 2021 Further, majority of the non-platinum drugs except irinotecan increased ERK1/2 activation in platinum-taxol resistant cells as observed by live-cell BRET assessment which were associated with p90RSK1/2 and BAD activation along with upregulation of multidrug transporter gene ABCC1 and cell survival genes like cyclin D1 and Bcl2. Irinotecan 51-61 ribosomal protein S6 kinase A1 Homo sapiens 192-201 34365218-5 2021 Further, majority of the non-platinum drugs except irinotecan increased ERK1/2 activation in platinum-taxol resistant cells as observed by live-cell BRET assessment which were associated with p90RSK1/2 and BAD activation along with upregulation of multidrug transporter gene ABCC1 and cell survival genes like cyclin D1 and Bcl2. Irinotecan 51-61 ATP binding cassette subfamily C member 1 Homo sapiens 275-280 34365218-5 2021 Further, majority of the non-platinum drugs except irinotecan increased ERK1/2 activation in platinum-taxol resistant cells as observed by live-cell BRET assessment which were associated with p90RSK1/2 and BAD activation along with upregulation of multidrug transporter gene ABCC1 and cell survival genes like cyclin D1 and Bcl2. Irinotecan 51-61 cyclin D1 Homo sapiens 310-319 34365218-5 2021 Further, majority of the non-platinum drugs except irinotecan increased ERK1/2 activation in platinum-taxol resistant cells as observed by live-cell BRET assessment which were associated with p90RSK1/2 and BAD activation along with upregulation of multidrug transporter gene ABCC1 and cell survival genes like cyclin D1 and Bcl2. Irinotecan 51-61 BCL2 apoptosis regulator Homo sapiens 324-328 34703007-6 2022 SN-38 clearance was influenced by rs887829 in UGT1A1, pre-treatment total bilirubin, and EGFR rare variant burden. Irinotecan 0-5 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 46-52 34703007-6 2022 SN-38 clearance was influenced by rs887829 in UGT1A1, pre-treatment total bilirubin, and EGFR rare variant burden. Irinotecan 0-5 epidermal growth factor receptor Homo sapiens 89-93 34703007-7 2022 Within each UGT1A1 genotype group, elevated pre-treatment total bilirubin and/or presence of at least one rare variant in EGFR resulted in significantly lower SN-38 clearance. Irinotecan 159-164 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-18 34703007-7 2022 Within each UGT1A1 genotype group, elevated pre-treatment total bilirubin and/or presence of at least one rare variant in EGFR resulted in significantly lower SN-38 clearance. Irinotecan 159-164 epidermal growth factor receptor Homo sapiens 122-126 34216140-9 2021 Mice injected with irinotecan presented systemic bacterial translocation and increased TLR4 immunostaining in the intestine. Irinotecan 19-29 toll-like receptor 4 Mus musculus 87-91 34680166-4 2021 The major constituent in this fraction was isolated and found to be 5,3",5"-trihydroxy-3,6,7,4"-tetramethoxyflavone (2), which at a concentration of 25 mug/mL potentiated the growth-inhibitory activity of SN-38 to a degree comparable to that of the known BCRP inhibitor Ko143 at 1 muM. Irinotecan 205-210 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 255-259 34680166-5 2021 A dye accumulation experiment suggested that 2 inhibits BCRP, and docking studies showed that 2 binds to the same BCRP site as SN-38. Irinotecan 127-132 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 56-60 34680166-5 2021 A dye accumulation experiment suggested that 2 inhibits BCRP, and docking studies showed that 2 binds to the same BCRP site as SN-38. Irinotecan 127-132 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 114-118 34680166-6 2021 These results indicate that 2 acts synergistically with SN-38, with 2 being a BCRP efflux pump inhibitor while SN-38 inhibits topoisomerase-1. Irinotecan 56-61 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 78-82 34216140-10 2021 In line with the clinical findings, Tlr4 gene deficiency enhanced irinotecan-related diarrhea and TLR9 expression in mice. Irinotecan 66-76 toll-like receptor 4 Mus musculus 36-40 34216140-11 2021 An increased myeloperoxidase activity and Il-18 expression (2.5-fold change) along with IL-10 decreased production in Tlr4-/- mice (P<0.05 vs. irinotecan-treated WT group) indicated boosted intestinal damage and inflammatory response. Irinotecan 143-153 myeloperoxidase Mus musculus 13-28 34216140-11 2021 An increased myeloperoxidase activity and Il-18 expression (2.5-fold change) along with IL-10 decreased production in Tlr4-/- mice (P<0.05 vs. irinotecan-treated WT group) indicated boosted intestinal damage and inflammatory response. Irinotecan 143-153 interleukin 18 Mus musculus 42-47 34216140-11 2021 An increased myeloperoxidase activity and Il-18 expression (2.5-fold change) along with IL-10 decreased production in Tlr4-/- mice (P<0.05 vs. irinotecan-treated WT group) indicated boosted intestinal damage and inflammatory response. Irinotecan 143-153 interleukin 10 Mus musculus 88-93 34486919-4 2021 Furthermore, prospective trials have shown that patients harboring the UGT1A1 *1/*1 and *1/*28 genotypes can tolerate higher doses of irinotecan, which may in turn impact on a better outcome. Irinotecan 134-144 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 71-77 34486919-5 2021 Upfront UGT1A1 genotyping could therefore be a usefulness strategy in order to individualize irinotecan dosing, but consensus on the recommended dose based on the UGT1A1 genotype is still lacking. Irinotecan 93-103 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 8-14 34486919-8 2021 EXPERT OPINION: Implementation of UGT1A1*28 and UGT1A1*6 genotyping in clinical practice is a first step toward personalizing irinotecan therapy. Irinotecan 126-136 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 34-40 34486919-8 2021 EXPERT OPINION: Implementation of UGT1A1*28 and UGT1A1*6 genotyping in clinical practice is a first step toward personalizing irinotecan therapy. Irinotecan 126-136 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 48-54 34176192-8 2021 IMPLICATIONS FOR PRACTICE: Sacituzumab govitecan (TRODELVY) is a novel antibody-drug conjugate composed of the active metabolite of irinotecan (SN-38) conjugated to a monoclonal antibody targeting Trop-2, an epithelial cell surface antigen overexpressed in many cancers. Irinotecan 144-149 tumor associated calcium signal transducer 2 Homo sapiens 197-203 34641401-5 2021 When synergy was observed, for example with curcumin and irinotecan, this was unrelated to MET induction, as assessed by changes in E-cadherin and vimentin expression. Irinotecan 57-67 cadherin 1 Homo sapiens 132-142 34176192-2 2021 Sacituzumab govitecan (IMMU-132) is an antibody-drug conjugate that contains the irinotecan active metabolite, SN-38, linked to a humanized monoclonal antibody targeting trophoblast cell surface antigen 2 (Trop-2), which is overexpressed in many solid tumors. Irinotecan 111-116 tumor associated calcium signal transducer 2 Homo sapiens 206-212 34176192-8 2021 IMPLICATIONS FOR PRACTICE: Sacituzumab govitecan (TRODELVY) is a novel antibody-drug conjugate composed of the active metabolite of irinotecan (SN-38) conjugated to a monoclonal antibody targeting Trop-2, an epithelial cell surface antigen overexpressed in many cancers. Irinotecan 132-142 tumor associated calcium signal transducer 2 Homo sapiens 197-203 34528449-3 2021 Results: A novel cis-eQTL, rs28967009, was identified for UGT1A1, which is predicted to upregulate UGT1A1 expression thereby potentially affecting the metabolism of dolutegravir and irinotecan, which are extensively prescribed in SA for HIV and colorectal cancer treatment, respectively. Irinotecan 182-192 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 58-64 34528449-3 2021 Results: A novel cis-eQTL, rs28967009, was identified for UGT1A1, which is predicted to upregulate UGT1A1 expression thereby potentially affecting the metabolism of dolutegravir and irinotecan, which are extensively prescribed in SA for HIV and colorectal cancer treatment, respectively. Irinotecan 182-192 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 99-105 34528449-4 2021 Conclusion: As increased UGT1A1 expression could affect the clinical outcome of dolutegravir and irinotecan treatment by increasing drug clearance, patients with the rs28967009A variant may require increased drug doses to reach therapeutic levels or should be prescribed alternative drugs. Irinotecan 97-107 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 25-31 34641401-5 2021 When synergy was observed, for example with curcumin and irinotecan, this was unrelated to MET induction, as assessed by changes in E-cadherin and vimentin expression. Irinotecan 57-67 vimentin Homo sapiens 147-155 34504265-9 2021 Both irinotecan and SN38 led to a global downregulation of genes involved in fibrosis such as COL1A1, COL1A2, MMP1 and ACTA2 in dermal fibroblasts from SSc patients (respectively - 27; - 20.5; - 30.2 and - 30% for irinotecan and - 61; - 55; - 50 and - 54% for SN38). Irinotecan 5-15 collagen type I alpha 1 chain Homo sapiens 94-100 34572902-5 2021 VKNG-1, at 6 microM, selectively inhibited ABCG2 transporter and sensitized ABCG2-overexpressing drug-resistant cancer cells to the ABCG2 substrate anticancer drugs mitoxantrone, SN-38, and doxorubicin in ABCG2-overexpressing colon cancers. Irinotecan 179-184 ATP binding cassette subfamily G member 2 (Junior blood group) Mus musculus 76-81 34572902-5 2021 VKNG-1, at 6 microM, selectively inhibited ABCG2 transporter and sensitized ABCG2-overexpressing drug-resistant cancer cells to the ABCG2 substrate anticancer drugs mitoxantrone, SN-38, and doxorubicin in ABCG2-overexpressing colon cancers. Irinotecan 179-184 ATP binding cassette subfamily G member 2 (Junior blood group) Mus musculus 132-137 34572902-9 2021 In addition, VKNG-1 enhanced the anticancer efficacy of irinotecan in ABCG2- overexpressing mouse tumor xenografts. Irinotecan 56-66 ATP binding cassette subfamily G member 2 (Junior blood group) Mus musculus 70-75 34504265-9 2021 Both irinotecan and SN38 led to a global downregulation of genes involved in fibrosis such as COL1A1, COL1A2, MMP1 and ACTA2 in dermal fibroblasts from SSc patients (respectively - 27; - 20.5; - 30.2 and - 30% for irinotecan and - 61; - 55; - 50 and - 54% for SN38). Irinotecan 5-15 collagen type I alpha 2 chain Homo sapiens 102-108 34504265-9 2021 Both irinotecan and SN38 led to a global downregulation of genes involved in fibrosis such as COL1A1, COL1A2, MMP1 and ACTA2 in dermal fibroblasts from SSc patients (respectively - 27; - 20.5; - 30.2 and - 30% for irinotecan and - 61; - 55; - 50 and - 54% for SN38). Irinotecan 5-15 matrix metallopeptidase 1 Homo sapiens 110-114 34504265-9 2021 Both irinotecan and SN38 led to a global downregulation of genes involved in fibrosis such as COL1A1, COL1A2, MMP1 and ACTA2 in dermal fibroblasts from SSc patients (respectively - 27; - 20.5; - 30.2 and - 30% for irinotecan and - 61; - 55; - 50 and - 54% for SN38). Irinotecan 5-15 actin alpha 2, smooth muscle Homo sapiens 119-124 34572750-9 2021 Nine patients with toxicity-associated pharmacogenomic variants were treated with a relevant medication: seven UGT1A1 intermediate metabolizers were treated with irinotecan, one intermediate DPYD metabolizer was treated with 5-fluorouracil, and one TPMT poor metabolizer was treated with mercaptopurine. Irinotecan 162-172 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 111-117 34572750-11 2021 One UGT1A1 heterozygote died after a single dose of irinotecan due to irinotecan-related adverse effects. Irinotecan 52-62 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 34337875-8 2021 The level of AURKA was significantly associated with the resistance to SB 505124, NU-7441, and irinotecan drugs (p < 0.01). Irinotecan 95-105 aurora kinase A Homo sapiens 13-18 34572750-11 2021 One UGT1A1 heterozygote died after a single dose of irinotecan due to irinotecan-related adverse effects. Irinotecan 70-80 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 34116144-1 2021 BACKGROUND: The pivotal phase III ASCENT trial demonstrated improved survival outcomes associated with sacituzumab govitecan (SG), an anti-Trop-2 antibody-drug conjugate linked with the topoisomerase-inhibitor SN-38, over single-agent chemotherapy of physician"s choice (TPC) in previously-treated metastatic triple-negative breast cancer (mTNBC). Irinotecan 210-215 tumor associated calcium signal transducer 2 Homo sapiens 139-145 34117512-1 2021 PURPOSE: SN-38, a pharmacologically active metabolite of irinotecan, is taken up into hepatocytes by organic anion transporting polypeptide (OATP) 1B1. Irinotecan 9-14 solute carrier organic anion transporter family member 1B1 Homo sapiens 101-150 34117512-1 2021 PURPOSE: SN-38, a pharmacologically active metabolite of irinotecan, is taken up into hepatocytes by organic anion transporting polypeptide (OATP) 1B1. Irinotecan 57-67 solute carrier organic anion transporter family member 1B1 Homo sapiens 101-150 34117512-2 2021 The effects of functional OATP1B1 521T>C on the pharmacokinetics of SN-38 remain controversial. Irinotecan 68-73 solute carrier organic anion transporter family member 1B1 Homo sapiens 26-33 34117512-3 2021 Here, we prospectively examined the effects of OATP1B1 function on the area under the plasma total or unbound concentration-time curve (tAUC or uAUC) of SN-38 by assessing OATP1B1 521T>C and the plasma levels of endogenous OATP1B1 substrates, coproporphyrin (CP)-I and III, in cancer patients treated with irinotecan. Irinotecan 153-158 solute carrier organic anion transporter family member 1B1 Homo sapiens 47-54 34117512-3 2021 Here, we prospectively examined the effects of OATP1B1 function on the area under the plasma total or unbound concentration-time curve (tAUC or uAUC) of SN-38 by assessing OATP1B1 521T>C and the plasma levels of endogenous OATP1B1 substrates, coproporphyrin (CP)-I and III, in cancer patients treated with irinotecan. Irinotecan 153-158 solute carrier organic anion transporter family member 1B1 Homo sapiens 172-179 34117512-12 2021 CONCLUSION: The contribution of OATP1B1 activity to inter-patient variability in the systemic exposure to SN-38 is likely minimal in patients without severe renal failure. Irinotecan 106-111 solute carrier organic anion transporter family member 1B1 Homo sapiens 32-39 34502348-4 2021 In this study, we discovered that by attenuating the drug transport function of ABCG2, TP-3654 resensitizes ABCG2-overexpressing multidrug-resistant cancer cells to cytotoxic ABCG2 substrate drugs topotecan, SN-38 and mitoxantrone. Irinotecan 208-213 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 80-85 34502348-4 2021 In this study, we discovered that by attenuating the drug transport function of ABCG2, TP-3654 resensitizes ABCG2-overexpressing multidrug-resistant cancer cells to cytotoxic ABCG2 substrate drugs topotecan, SN-38 and mitoxantrone. Irinotecan 208-213 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 108-113 34526906-0 2021 Corrigendum: 7-Ethyl-10-Hydroxycamptothecin, A DNA Topoisomerase I Inhibitor, Performs BRD4 Inhibitory Activity and Inhibits Human Leukemic Cell Growth. Irinotecan 13-43 bromodomain containing 4 Homo sapiens 87-91 34502348-4 2021 In this study, we discovered that by attenuating the drug transport function of ABCG2, TP-3654 resensitizes ABCG2-overexpressing multidrug-resistant cancer cells to cytotoxic ABCG2 substrate drugs topotecan, SN-38 and mitoxantrone. Irinotecan 208-213 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 175-180 34408079-6 2021 We demonstrated that induction of telomere dysfunction resulted in ATM activation that, in turn, conferred resistance to temozolomide + SN-38 (4.2-fold change in IC50, P < 0.001). Irinotecan 136-141 ataxia telangiectasia mutated Mus musculus 67-70 34408079-7 2021 ATM knockdown (shRNA) or inhibition using a clinical-stage small-molecule inhibitor (AZD0156) reversed resistance to temozolomide + irinotecan in ALT neuroblastoma cell lines in vitro (P < 0.001) and in four ALT xenografts in vivo (EFS, P < 0.0001). Irinotecan 132-142 ataxia telangiectasia mutated Mus musculus 0-3 34281853-3 2021 PATIENTS AND METHODS: We retrospectively collected the clinical data of KRAS exon 2 wild type (WT) mCRC patients treated with EGFR inhibitor monotherapy or EGFR inhibitor plus irinotecan as third-line chemotherapy. Irinotecan 176-186 KRAS proto-oncogene, GTPase Homo sapiens 72-76 34300575-6 2021 All three TDP1 inhibitors had no effect on TOP1 activity and acted synergistically with the TOP1 poison SN-38 to increase the amount of TOP1 cleavage-induced DNA damage. Irinotecan 104-109 tyrosyl-DNA phosphodiesterase 1 Homo sapiens 10-14 34300575-6 2021 All three TDP1 inhibitors had no effect on TOP1 activity and acted synergistically with the TOP1 poison SN-38 to increase the amount of TOP1 cleavage-induced DNA damage. Irinotecan 104-109 DNA topoisomerase I Homo sapiens 92-96 34300575-6 2021 All three TDP1 inhibitors had no effect on TOP1 activity and acted synergistically with the TOP1 poison SN-38 to increase the amount of TOP1 cleavage-induced DNA damage. Irinotecan 104-109 DNA topoisomerase I Homo sapiens 136-140 34300575-7 2021 Further, they promoted cell death even with low dose SN-38, thereby establishing two new classes of TDP1 inhibitors with clinical potential. Irinotecan 53-58 tyrosyl-DNA phosphodiesterase 1 Homo sapiens 100-104 34201500-8 2021 Interestingly, shortly after engraftment, Irinotecan completely blocked leukemia expansion in mouse xenografts of a pediatric MLL-rearranged ALL cell line, as well as in two patient-derived xenograft (PDX) models of MLL-rearranged infant ALL. Irinotecan 42-52 lysine (K)-specific methyltransferase 2A Mus musculus 126-129 34193275-2 2021 Recently, we have reported that CPT-11-induced gastrointestinal damage is associated with the upregulation of intestinal P-glycoprotein (P-gp) expression and decreased absorption of its substrate, dabigatran etexilate (DABE), using a rat model. Irinotecan 32-38 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 121-135 34193275-2 2021 Recently, we have reported that CPT-11-induced gastrointestinal damage is associated with the upregulation of intestinal P-glycoprotein (P-gp) expression and decreased absorption of its substrate, dabigatran etexilate (DABE), using a rat model. Irinotecan 32-38 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 137-141 34193275-4 2021 The aim of this study was to quantitatively evaluate how P-gp activity changes in rats with CPT-11-induced gastrointestinal damage, as assessed by the absorption of digoxin (DGX), a typical P-gp substrate. Irinotecan 92-98 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 57-61 34193275-4 2021 The aim of this study was to quantitatively evaluate how P-gp activity changes in rats with CPT-11-induced gastrointestinal damage, as assessed by the absorption of digoxin (DGX), a typical P-gp substrate. Irinotecan 92-98 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 190-194 34193275-11 2021 CONCLUSIONS: Increased P-gp expression in rats with CPT-11-induced gastrointestinal damage is not necessarily associated with increased P-gp activity or contribution to the drug absorption in vivo. Irinotecan 52-58 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 23-27 34201500-9 2021 Also, from a more clinically relevant perspective, Irinotecan monotherapy was able to induce sustainable disease remissions in MLL-rearranged ALL xenotransplanted mice burdened with advanced leukemia. Irinotecan 51-61 lysine (K)-specific methyltransferase 2A Mus musculus 127-130 34201500-10 2021 Taken together, our data demonstrate that Irinotecan exerts highly potent anti-leukemia effects against pediatric MLL-rearranged ALL, and likely against other, more favorable subtypes of childhood ALL as well. Irinotecan 42-52 lysine (K)-specific methyltransferase 2A Mus musculus 114-117 34212047-10 2021 Results: FXYD6 was downregulated in CRC resistant patients and irinotecan- (Iri-) resistant SW620 cells (SW620/Iri). Irinotecan 63-73 FXYD domain containing ion transport regulator 6 Homo sapiens 9-14 34212047-11 2021 FXYD6 silence inhibited cell apoptosis and enhanced prosurvival autophagy, whereas FXYD6 overexpression produced the opposite effect which alleviated the drug resistance to irinotecan and oxaliplatin of CRC cells. Irinotecan 173-183 FXYD domain containing ion transport regulator 6 Homo sapiens 0-5 34212047-11 2021 FXYD6 silence inhibited cell apoptosis and enhanced prosurvival autophagy, whereas FXYD6 overexpression produced the opposite effect which alleviated the drug resistance to irinotecan and oxaliplatin of CRC cells. Irinotecan 173-183 FXYD domain containing ion transport regulator 6 Homo sapiens 83-88 34201243-3 2021 The aim of this study was to evaluate the repositioning of the anti-cancer molecule irinotecan as a potential modulator of the antiviral and inflammatory responses of primary human synovial fibroblasts (HSF), the main stromal cells of the joint synovium. Irinotecan 84-94 interleukin 6 Homo sapiens 203-206 34201243-6 2021 Quantitative RT-PCR analysis revealed that irinotecan at 15 microM was able to amplify the antiviral response (i.e., interferon-stimulated gene expression) of HSF exposed to PIC and reduce the expression of pro-inflammatory genes (CXCL8, IL-6 and COX-2) upon IL-1beta treatment. Irinotecan 43-53 interleukin 6 Homo sapiens 159-162 34201243-6 2021 Quantitative RT-PCR analysis revealed that irinotecan at 15 microM was able to amplify the antiviral response (i.e., interferon-stimulated gene expression) of HSF exposed to PIC and reduce the expression of pro-inflammatory genes (CXCL8, IL-6 and COX-2) upon IL-1beta treatment. Irinotecan 43-53 C-X-C motif chemokine ligand 8 Homo sapiens 231-236 34201243-6 2021 Quantitative RT-PCR analysis revealed that irinotecan at 15 microM was able to amplify the antiviral response (i.e., interferon-stimulated gene expression) of HSF exposed to PIC and reduce the expression of pro-inflammatory genes (CXCL8, IL-6 and COX-2) upon IL-1beta treatment. Irinotecan 43-53 interleukin 6 Homo sapiens 238-242 34201243-6 2021 Quantitative RT-PCR analysis revealed that irinotecan at 15 microM was able to amplify the antiviral response (i.e., interferon-stimulated gene expression) of HSF exposed to PIC and reduce the expression of pro-inflammatory genes (CXCL8, IL-6 and COX-2) upon IL-1beta treatment. Irinotecan 43-53 mitochondrially encoded cytochrome c oxidase II Homo sapiens 247-252 34201243-6 2021 Quantitative RT-PCR analysis revealed that irinotecan at 15 microM was able to amplify the antiviral response (i.e., interferon-stimulated gene expression) of HSF exposed to PIC and reduce the expression of pro-inflammatory genes (CXCL8, IL-6 and COX-2) upon IL-1beta treatment. Irinotecan 43-53 interleukin 1 alpha Homo sapiens 259-267 34268956-1 2021 PURPOSE: The purpose of this study was to compare the clinical efficacy and safety of S-1 + oxaliplatin (SOX) chemotherapy regimen combined with trastuzumab and irinotecan + cisplatin (IP) chemotherapy regimen combined with trastuzumab in treating human epidermal growth factor receptor 2 (HER-2)-positive advanced gastric cancer. Irinotecan 161-171 erb-b2 receptor tyrosine kinase 2 Homo sapiens 254-288 34084136-1 2021 Human carboxylesterase 2 (CES2), one of the most abundant hydrolases distributed in the small intestine, has been validated as a key therapeutic target to ameliorate the intestinal toxicity caused by irinotecan. Irinotecan 200-210 carboxylesterase 2 Homo sapiens 6-24 34268956-1 2021 PURPOSE: The purpose of this study was to compare the clinical efficacy and safety of S-1 + oxaliplatin (SOX) chemotherapy regimen combined with trastuzumab and irinotecan + cisplatin (IP) chemotherapy regimen combined with trastuzumab in treating human epidermal growth factor receptor 2 (HER-2)-positive advanced gastric cancer. Irinotecan 161-171 erb-b2 receptor tyrosine kinase 2 Homo sapiens 290-295 35415893-2 2022 The aim of this clinical study was to evaluate the safety, efficacy and pharmacokinetics of a novel regimen comprised of metronomic oxaliplatin (O), chronomodulated capecitabine (X) and UGT1A1 genotype-guided dosing of irinotecan (IRI) (OXIRI) as well as its immunomodulatory effects. Irinotecan 219-229 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 186-192 35415893-7 2022 UGT1A1-genotype directed dosing resulted in similar exposure levels of irinotecan, SN-38 and SN-38G in all patients. Irinotecan 71-81 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 35415893-7 2022 UGT1A1-genotype directed dosing resulted in similar exposure levels of irinotecan, SN-38 and SN-38G in all patients. Irinotecan 83-88 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 35579261-0 2022 Targeted delivery of irinotecan and SLP2 shRNA with GRP-conjugated magnetic graphene oxide for glioblastoma treatment. Irinotecan 21-31 gastrin releasing peptide Homo sapiens 52-55 35624189-4 2022 In this research, MMAE, SN-38, and DXd were selected as candidates for payloads of the anti-Trop-2 mAb SY02. Irinotecan 24-29 tumor associated calcium signal transducer 2 Homo sapiens 92-98 35430260-9 2022 We identified that bile excretion and UDP-glucuronosyltransferases (UGT) played more important roles than fecal excretion and renal clearance in SN-38 pharmacokinetics. Irinotecan 145-150 beta-1,3-glucuronyltransferase 2 (glucuronosyltransferase S) Mus musculus 38-66 35430260-9 2022 We identified that bile excretion and UDP-glucuronosyltransferases (UGT) played more important roles than fecal excretion and renal clearance in SN-38 pharmacokinetics. Irinotecan 145-150 beta-1,3-glucuronyltransferase 2 (glucuronosyltransferase S) Mus musculus 68-71 35637361-1 2022 BACKGROUND: UGT1A1 polymorphisms should be considered when using irinotecan-containing regimens, especially in patients with a double-variant-type (DV), including homozygous for UGT1A1*28 and UGT1A1*6 and heterozygous for both UGT1A1*28 and UGT1A1*6. Irinotecan 65-75 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-18 35545724-1 2022 PURPOSE: Sacituzumab govitecan (SG) is an antibody-drug conjugate composed of an anti-Trop-2 antibody coupled to SN-38 via a proprietary hydrolyzable linker. Irinotecan 113-118 tumor associated calcium signal transducer 2 Homo sapiens 86-92 35506159-0 2022 Association of UGT1A1*6,*28 or ABCC2 c.3972C>T genetic polymorphisms with irinotecan induced toxicity in Asian cancer patients: Meta-analysis. Irinotecan 74-84 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 15-21 35177165-1 2022 Uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) polymorphism plays a crucial role in the increased susceptibility and toxicity of patients to irinotecan. Irinotecan 151-161 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-47 35177165-1 2022 Uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) polymorphism plays a crucial role in the increased susceptibility and toxicity of patients to irinotecan. Irinotecan 151-161 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 49-55 35177165-2 2022 This retrospective, observational study compared the clinical outcomes and adverse events (AEs) in RAS wild-type metastatic colorectal cancer (mCRC) patients treated with cetuximab or bevacizumab plus FOLFIRI with UGT1A1 genotyping and irinotecan dose escalation as the first-line therapy. Irinotecan 236-246 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 214-220 35177165-5 2022 All patients received irinotecan dose escalation based on UGT1A1 genotyping. Irinotecan 22-32 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 58-64 35177165-10 2022 Our results revealed that patients with wild-type RAS mCRC, regardless of biologics, with UGT1A1 genotyping can tolerate escalated doses of irinotecan and potentially achieve a more favorable clinical outcome without significantly increased toxicity. Irinotecan 140-150 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 90-96 35628140-5 2022 Using a unique transgenic mouse (FAT-1) model combined with dietary supplementation experiments, we demonstrate that an elevated tissue n-3 PUFA status with a decreased n-6/n-3 PUFA ratio significantly reduces CPT-11-induced weight loss, bloody diarrhea, gut pathological changes, and mortality. Irinotecan 210-216 FAT atypical cadherin 1 Mus musculus 33-38 35506159-0 2022 Association of UGT1A1*6,*28 or ABCC2 c.3972C>T genetic polymorphisms with irinotecan induced toxicity in Asian cancer patients: Meta-analysis. Irinotecan 74-84 ATP binding cassette subfamily C member 2 Homo sapiens 31-36 35506159-2 2022 Also, ABCC2 c.3972C>T may affect toxicity of irinotecan. Irinotecan 45-55 ATP binding cassette subfamily C member 2 Homo sapiens 6-11 35592314-16 2022 Finally, specific inhibitors, like irinotecan and puromycin, which correlate with SPA17 expression in different cancer types, were also screened using Connectivity Map (CMap). Irinotecan 35-45 sperm autoantigenic protein 17 Homo sapiens 82-87 35506159-3 2022 It was aimed to assess the aggregated risk of neutropenia or diarrhea in Asian cancer patients taking irinotecan and inherited UGT1A1*6, UGT1A1*28 or ABCC2 c.3972C>T genetic variants. Irinotecan 102-112 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 137-143 35506159-3 2022 It was aimed to assess the aggregated risk of neutropenia or diarrhea in Asian cancer patients taking irinotecan and inherited UGT1A1*6, UGT1A1*28 or ABCC2 c.3972C>T genetic variants. Irinotecan 102-112 ATP binding cassette subfamily C member 2 Homo sapiens 150-155 35506159-9 2022 Both UGT1A1*6 and UGT1A1*28 genetic variants should be screened in Asian cancer patients to reduce substantially irinotecan induced severe toxicities. Irinotecan 113-123 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 5-11 35506159-9 2022 Both UGT1A1*6 and UGT1A1*28 genetic variants should be screened in Asian cancer patients to reduce substantially irinotecan induced severe toxicities. Irinotecan 113-123 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 18-24 35530278-4 2022 SG consists of an anti-trophoblast cell-surface antigen 2 (Trop2) antibody conjugated with a topoisomerase I inhibitor, SN-38, which is diffused out of the targeted Trop2 positive cancer cells and induces the bystander killing effect on surrounding cells regardless of their Trop2 expression status. Irinotecan 120-125 tumor associated calcium signal transducer 2 Homo sapiens 59-64 35489740-2 2022 Therefore, we conducted a phase II study to verify the efficacy and safety of the combination of S-1 and irinotecan plus bevacizumab (SIRB regimen) as second-line treatment for patients with oxaliplatin and cetuximab-refractory KRAS wild-type mCRC. Irinotecan 105-115 KRAS proto-oncogene, GTPase Homo sapiens 228-232 35233973-5 2022 We also assessed the impact of UGT1A1 gene polymorphisms, which are involved in irinotecan metabolism, on outcomes and toxicity. Irinotecan 80-90 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 31-37 35147853-0 2022 Prediction of Drug-Drug Interaction Between Dabrafenib and Irinotecan via UGT1A1-Mediated Glucuronidation. Irinotecan 59-69 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 74-80 35147853-4 2022 METHODS: Recombinant human uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) and human liver microsomes (HLMs) were employed to catalyze the glucuronidation of SN-38 in vitro. Irinotecan 167-172 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 27-74 35147853-4 2022 METHODS: Recombinant human uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) and human liver microsomes (HLMs) were employed to catalyze the glucuronidation of SN-38 in vitro. Irinotecan 167-172 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 76-82 35147853-6 2022 RESULTS: Dabrafenib noncompetitively inhibited SN-38 glucuronidation in pooled HLMs and recombinant UGT1A1 with unbound inhibitor constant (Ki,u) values of 12.43 +- 0.28 and 3.89 +- 0.40 muM, respectively. Irinotecan 47-52 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 100-106 35147853-9 2022 CONCLUSION: Dabrafenib is a potent noncompetitive inhibitor of UGT1A1 and may bring potential risk of DDI when combined with irinotecan. Irinotecan 125-135 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 63-69 35498511-8 2022 All patients with PC-only CRC were treated with first-line bevacizumab and FOLFIRI, and the irinotecan dose escalation depended on UGT1A1 polymorphism. Irinotecan 92-102 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 131-137 35498511-13 2022 Conclusion: Our study revealed that in patients with PC-only mCRC treatment of first-line bevacizumab and FOLFIRI through irinotecan dose escalation according to UGT1A1 polymorphism could confer such patients with comparable outcomes to that of patients with LiM-only and LuM-only mCRC. Irinotecan 122-132 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 162-168 35354375-8 2022 G-CSF use was observed among 63.6%, 34.9%, 33.9%, and 44.9% of patients treated with FOLFIRINOX, FOLFOX, FOLFIRI, and liposomal irinotecan-based regimens, respectively. Irinotecan 128-138 colony stimulating factor 3 Homo sapiens 0-5 35445479-6 2022 SN-38 is inactivated by UGT1A1 enzymes. Irinotecan 0-5 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 24-30 35530278-4 2022 SG consists of an anti-trophoblast cell-surface antigen 2 (Trop2) antibody conjugated with a topoisomerase I inhibitor, SN-38, which is diffused out of the targeted Trop2 positive cancer cells and induces the bystander killing effect on surrounding cells regardless of their Trop2 expression status. Irinotecan 120-125 tumor associated calcium signal transducer 2 Homo sapiens 165-170 35530278-4 2022 SG consists of an anti-trophoblast cell-surface antigen 2 (Trop2) antibody conjugated with a topoisomerase I inhibitor, SN-38, which is diffused out of the targeted Trop2 positive cancer cells and induces the bystander killing effect on surrounding cells regardless of their Trop2 expression status. Irinotecan 120-125 tumor associated calcium signal transducer 2 Homo sapiens 275-280 35231830-5 2022 Moreover, we firstly proved that ATM inhibitors could sensitize Irinotecan and Etoposide in a time-dependent manner on MCF-7 and SW480 cells, antagonism in a short period treatment while synergy at a long-term treatment and ATM agonist worked in an opposite way of ATM inhibitors. Irinotecan 64-74 ATM serine/threonine kinase Homo sapiens 33-36 35412430-3 2022 Research suggests an association between methylated DCR1 (mDCR1) and lack of benefit with irinotecan (IFL) treatment. Irinotecan 90-100 dicer 1, ribonuclease III Homo sapiens 52-56 35396702-9 2022 The findings of this study suggest that irinotecan-induced hepatotoxicity is not directly associated with genetic variables but is mostly related to concomitant use of CYP3A inducers and platinum, as well as the presence of liver metastasis and CVD. Irinotecan 40-50 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 168-173 35231830-5 2022 Moreover, we firstly proved that ATM inhibitors could sensitize Irinotecan and Etoposide in a time-dependent manner on MCF-7 and SW480 cells, antagonism in a short period treatment while synergy at a long-term treatment and ATM agonist worked in an opposite way of ATM inhibitors. Irinotecan 64-74 ATM serine/threonine kinase Homo sapiens 224-227 35231830-5 2022 Moreover, we firstly proved that ATM inhibitors could sensitize Irinotecan and Etoposide in a time-dependent manner on MCF-7 and SW480 cells, antagonism in a short period treatment while synergy at a long-term treatment and ATM agonist worked in an opposite way of ATM inhibitors. Irinotecan 64-74 ATM serine/threonine kinase Homo sapiens 265-268 35240418-6 2022 Co-treatment by CPT-11 and 10o significantly diminished the tumor volume, indicating the great potential of 10o as a candidate of Chk1/2 inhibitor. Irinotecan 16-22 checkpoint kinase 1 Mus musculus 130-136 35187743-0 2022 eIF3a-PPP2R5A-mediated ATM/ATR dephosphorylation is essential for irinotecan-induced DNA damage response. Irinotecan 66-76 eukaryotic translation initiation factor 3 subunit J Homo sapiens 0-5 35187743-0 2022 eIF3a-PPP2R5A-mediated ATM/ATR dephosphorylation is essential for irinotecan-induced DNA damage response. Irinotecan 66-76 protein phosphatase 2 regulatory subunit B'alpha Homo sapiens 6-13 35187743-0 2022 eIF3a-PPP2R5A-mediated ATM/ATR dephosphorylation is essential for irinotecan-induced DNA damage response. Irinotecan 66-76 ATM serine/threonine kinase Homo sapiens 23-26 35187743-0 2022 eIF3a-PPP2R5A-mediated ATM/ATR dephosphorylation is essential for irinotecan-induced DNA damage response. Irinotecan 66-76 ATR serine/threonine kinase Homo sapiens 27-30 35187743-3 2022 This study focused on the role of eIF3a in irinotecan-induced DNA damage response. Irinotecan 43-53 eukaryotic translation initiation factor 3 subunit J Homo sapiens 34-39 35187743-4 2022 MATERIALS AND METHODS: The cck8 cell viability and clone survival analyses were used to test the regulatory role of eIF3a on irinotecan sensitivity in HT29 and CACO2 cell lines in vitro. Irinotecan 125-135 eukaryotic translation initiation factor 3 subunit J Homo sapiens 116-121 35187743-9 2022 Suppression of PPP2R5A resulted in chronic ATM/ATR phosphorylation and activation, impairing DNA repair and enhancing irinotecan sensitivity. Irinotecan 118-128 protein phosphatase 2 regulatory subunit B'alpha Homo sapiens 15-22 35187743-10 2022 CONCLUSIONS: Our study suggested eIF3a with a high potential to influence phenotypic functions, which may contribute substantially to the early identification of susceptible individuals and the provision of personalized medication to irinotecan-treated patients. Irinotecan 234-244 eukaryotic translation initiation factor 3 subunit J Homo sapiens 33-38 35456602-7 2022 As demonstrated by the results of inhibition on multi-receptors by Sunitinib, we confirmed that SN-38/Sunitinib co-loaded micelles to be a treatment modality that could inhibit VEGF and PDGF receptors and enhance the antitumor effect of SN-38 (p < 0.05). Irinotecan 96-101 vascular endothelial growth factor A Homo sapiens 177-181 35061524-0 2022 Precision Medicine: UGT1A1 Genotyping to Better Manage Irinotecan-Induced Toxicity. Irinotecan 55-65 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 20-26 35190308-7 2022 We further found that bile acid mixture (BA mix) could inhibit hOAT2-mediated uptake of cGMP, 5-fluorouracil, irinotecan and paclitaxel. Irinotecan 110-120 solute carrier family 22 member 7 Homo sapiens 63-68 35481414-1 2022 OBJECTIVE: To undertake a meta-analysis of the treatment effects of different second-line chemotherapy regimens compared with FOLFIRINOX (FOL (folinic acid), F (fluorouracil), IRIN (irinotecan), OX (oxaliplatin)) after failure of gemcitabine-based first-line therapy in patients with pancreatic cancer. Irinotecan 176-180 hypocretin neuropeptide precursor Homo sapiens 195-197 35361811-6 2022 Using isogenic HCT116 clones, we showed that this mutation of ATR sensitizes the cells to several drugs, including SN-38 (topoisomerase I inhibitor) and VE-822 (ATR inhibitor) and exacerbates their synergistic effects. Irinotecan 115-120 ATR serine/threonine kinase Homo sapiens 62-65 35059735-0 2022 Targeting MUCL1 protein inhibits cell proliferation and EMT by deregulating beta-catenin and increases irinotecan sensitivity in colorectal cancer. Irinotecan 103-113 mucin like 1 Homo sapiens 10-15 35192728-4 2022 Together with extended tumor exposure to therapeutically effective drug levels via reversible conjugation to Pluronic F-108 (PF108), these features translated into rapid tumor regression and long-term survival in models of both ABCG2-overexpressing and p53-mutant high-risk neuroblastomas, in contrast to a marginal effect of the clinically used camptothecin derivative, irinotecan. Irinotecan 371-381 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 228-233 35192728-4 2022 Together with extended tumor exposure to therapeutically effective drug levels via reversible conjugation to Pluronic F-108 (PF108), these features translated into rapid tumor regression and long-term survival in models of both ABCG2-overexpressing and p53-mutant high-risk neuroblastomas, in contrast to a marginal effect of the clinically used camptothecin derivative, irinotecan. Irinotecan 371-381 tumor protein p53 Homo sapiens 253-256 35059735-14 2022 Targeting MUCL1 increases the drug sensitivity of CRC cells towards irinotecan. Irinotecan 68-78 mucin like 1 Homo sapiens 10-15 35226656-2 2022 Mutations in the UGT1A1 gene can be associated with increased toxicity from irinotecan-based chemotherapy. Irinotecan 76-86 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 17-23 35197630-7 2022 Our results show that irinotecan and CHEK1 inhibition have synergistic effects in microsatellite-stable or KRAS-TP53 double-mutant colon cancer cells, leading to apoptosis and suppression of tumour xenograft growth. Irinotecan 22-32 KRAS proto-oncogene, GTPase Homo sapiens 107-111 35197630-7 2022 Our results show that irinotecan and CHEK1 inhibition have synergistic effects in microsatellite-stable or KRAS-TP53 double-mutant colon cancer cells, leading to apoptosis and suppression of tumour xenograft growth. Irinotecan 22-32 tumor protein p53 Homo sapiens 112-116 35226656-5 2022 Genetic testing identified a mutation in the UGT1A1 gene associated with increased toxicity to irinotecan, and the EIA tests performed for evaluation of C. difficile toxins A and B showed repeatedly negative results. Irinotecan 95-105 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 45-51 35207692-1 2022 Current guidelines recommend pre-therapeutic UGT1A1 genotyping to guide irinotecan dosing, but the usefulness of this approach remains to be clarified. Irinotecan 72-82 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 45-51 35152824-0 2022 Mitochondrial DNA as a Possible Ligand for TLR9 in Irinotecan-induced Small Intestinal Mucositis. Irinotecan 51-61 toll-like receptor 9 Mus musculus 43-47 35152824-5 2022 WT mice injected with irinotecan presented a progressive increase in mtDNA in the serum along with increased hematocrit, shortening of small intestine length, reduction of intestinal villus:crypt ratio and increased influx of neutrophils, which were followed by higher expression of Nlrp3 and Casp1 mRNA and increased IL-1beta levels in the ileum when compared to vehicle-injected mice. Irinotecan 22-32 NLR family, pyrin domain containing 3 Mus musculus 283-288 35152824-5 2022 WT mice injected with irinotecan presented a progressive increase in mtDNA in the serum along with increased hematocrit, shortening of small intestine length, reduction of intestinal villus:crypt ratio and increased influx of neutrophils, which were followed by higher expression of Nlrp3 and Casp1 mRNA and increased IL-1beta levels in the ileum when compared to vehicle-injected mice. Irinotecan 22-32 caspase 1 Mus musculus 293-298 35152824-5 2022 WT mice injected with irinotecan presented a progressive increase in mtDNA in the serum along with increased hematocrit, shortening of small intestine length, reduction of intestinal villus:crypt ratio and increased influx of neutrophils, which were followed by higher expression of Nlrp3 and Casp1 mRNA and increased IL-1beta levels in the ileum when compared to vehicle-injected mice. Irinotecan 22-32 interleukin 1 alpha Mus musculus 318-326 35152824-8 2022 Overall, our findings show that the amount of circulating free CpG-DNA is increased upon chemotherapy and that TLR9 activation is important for NLRP3 inflammasome transcription and further IL-1beta release, playing a central role in the development of irinotecan-induced intestinal mucositis. Irinotecan 252-262 toll-like receptor 9 Mus musculus 111-115 35152824-8 2022 Overall, our findings show that the amount of circulating free CpG-DNA is increased upon chemotherapy and that TLR9 activation is important for NLRP3 inflammasome transcription and further IL-1beta release, playing a central role in the development of irinotecan-induced intestinal mucositis. Irinotecan 252-262 NLR family, pyrin domain containing 3 Mus musculus 144-149 35152824-8 2022 Overall, our findings show that the amount of circulating free CpG-DNA is increased upon chemotherapy and that TLR9 activation is important for NLRP3 inflammasome transcription and further IL-1beta release, playing a central role in the development of irinotecan-induced intestinal mucositis. Irinotecan 252-262 interleukin 1 alpha Mus musculus 189-197 35152824-9 2022 We suggest that TLR9 antagonism may be a new therapeutic strategy for limiting irinotecan-induced intestinal inflammation. Irinotecan 79-89 toll-like receptor 9 Mus musculus 16-20 35131032-4 2022 Our results also reveal that cells depleted of KChIP3 are four times more resistant (measured as cell viability and DNA damage) to chemotherapeutics 5-Fluorouracil plus Irinotecan (5-FU+iri.) Irinotecan 169-179 potassium voltage-gated channel interacting protein 3 Homo sapiens 47-53 34654938-3 2022 Patients with genotype UGT1A1*6 (exon 1, 211G > A) treated with the antineoplastic drug SN-38 have been reported to exhibit decreased glucuronide conjugation and increased incidence of intestinal toxicity and its severe side effects, including severe diarrhea. Irinotecan 88-93 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 23-29 34998046-0 2022 UGT1A1 genotype-guided dosing of irinotecan: A prospective safety and cost analysis in poor metaboliser patients. Irinotecan 33-43 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 34998046-1 2022 AIM: To determine the safety, feasibility, pharmacokinetics, and cost of UGT1A1 genotype-guided dosing of irinotecan. Irinotecan 106-116 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 73-79 34998046-2 2022 PATIENTS AND METHODS: In this prospective, multicentre, non-randomised study, patients intended for treatment with irinotecan were pre-therapeutically genotyped for UGT1A1*28 and UGT1A1*93. Irinotecan 115-125 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 165-171 34998046-2 2022 PATIENTS AND METHODS: In this prospective, multicentre, non-randomised study, patients intended for treatment with irinotecan were pre-therapeutically genotyped for UGT1A1*28 and UGT1A1*93. Irinotecan 115-125 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 179-185 34998046-9 2022 Systemic exposure of SN-38 of reduced dosing in UGT1A1 PMs was still slightly higher compared to a standard-dosed irinotecan patient cohort (difference: +32%). Irinotecan 21-26 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 48-54 34998046-11 2022 CONCLUSION: UGT1A1 genotype-guided dosing significantly reduces the incidence of febrile neutropenia in UGT1A1 PM patients treated with irinotecan, results in a therapeutically effective systemic drug exposure, and is cost-saving. Irinotecan 136-146 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-18 34998046-11 2022 CONCLUSION: UGT1A1 genotype-guided dosing significantly reduces the incidence of febrile neutropenia in UGT1A1 PM patients treated with irinotecan, results in a therapeutically effective systemic drug exposure, and is cost-saving. Irinotecan 136-146 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 104-110 34998046-12 2022 Therefore, UGT1A1 genotype-guided dosing of irinotecan should be considered standard of care in order to improve individual patient safety. Irinotecan 44-54 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 11-17 34654938-7 2022 The CYP3A4 POR UGT1A1*6 KI-Caco-2 cells were sensitive to SN-38-induced intestinal toxicity. Irinotecan 58-63 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 4-10 34654938-7 2022 The CYP3A4 POR UGT1A1*6 KI-Caco-2 cells were sensitive to SN-38-induced intestinal toxicity. Irinotecan 58-63 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 15-21 35127582-6 2022 Conclusion: Gemcitabine and L-OHP + EPI + irinotecan + 5-FU, L-OHP + EPI, and L-OHP + irinotecan + EPI were more effective against AFP-positive compared with AFP-negative liver cancer cells according to in vitro high-throughput drug sensitivity screening. Irinotecan 42-52 alpha fetoprotein Homo sapiens 158-161 35127582-6 2022 Conclusion: Gemcitabine and L-OHP + EPI + irinotecan + 5-FU, L-OHP + EPI, and L-OHP + irinotecan + EPI were more effective against AFP-positive compared with AFP-negative liver cancer cells according to in vitro high-throughput drug sensitivity screening. Irinotecan 86-96 alpha fetoprotein Homo sapiens 158-161 35049694-1 2022 The aim of this study was to examine the safety and efficacy of 40 microm and 75 microm calibrated irinotecan-eluting beads (DEBIRI-TACE) for the treatment of colorectal cancer metastases. Irinotecan 99-109 ADAM metallopeptidase domain 17 Homo sapiens 132-136 35070248-6 2022 A 44-year-old BRCA1-mutated ovarian cancer patient was treated in sixth line with lurbinectedin and irinotecan with a time to further progression (TTFP) equal to 8 months. Irinotecan 100-110 BRCA1 DNA repair associated Homo sapiens 14-19 35070248-0 2022 Exceptional response to lurbinectedin and irinotecan in BRCA-mutated platinum-resistant ovarian cancer patient: a case report. Irinotecan 42-52 BRCA1 DNA repair associated Homo sapiens 56-60 35070248-5 2022 This article describes an exceptional response to lurbinectedin alone followed by the association with irinotecan in a BRCA-mutated platinum-resistant ovarian cancer patient. Irinotecan 103-113 BRCA1 DNA repair associated Homo sapiens 119-123 35127384-4 2022 CPT-11 causes DNA damage and the release of double-stranded DNA (dsDNA) from the intestine, leading to cyclic-GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING)-mediated colitis, which was significantly decreased by andrographolide both in vivo and in vitro. Irinotecan 0-6 cyclic GMP-AMP synthase Mus musculus 103-126 35127384-4 2022 CPT-11 causes DNA damage and the release of double-stranded DNA (dsDNA) from the intestine, leading to cyclic-GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING)-mediated colitis, which was significantly decreased by andrographolide both in vivo and in vitro. Irinotecan 0-6 cyclic GMP-AMP synthase Mus musculus 128-132 35491733-4 2022 PC3 cells co-cultured with ADSC.CE.TRAIL showed higher cytotoxicity than did CPT-11 monotherapy, ADSC.CE, or ADSC.sTRAIL under CPT-11 treatment. Irinotecan 77-83 tumor necrosis factor (ligand) superfamily, member 10 Mus musculus 35-40 34996412-1 2022 BACKGROUND: Pharmacogenetic (PGx) testing for germline variants in the DPYD and UGT1A1 genes can be used to guide fluoropyrimidine and irinotecan dosing, respectively. Irinotecan 135-145 dihydropyrimidine dehydrogenase Homo sapiens 71-75 34996412-1 2022 BACKGROUND: Pharmacogenetic (PGx) testing for germline variants in the DPYD and UGT1A1 genes can be used to guide fluoropyrimidine and irinotecan dosing, respectively. Irinotecan 135-145 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 80-86 35356222-0 2022 UGT1A1 Allele Test Not Only Minimizes the Toxicity But Also Maximizes the Therapeutic Effect of Irinotecan in the Treatment of Colorectal Cancer: A Narrative Review. Irinotecan 96-106 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 35356222-2 2022 Adjusting the dose of irinotecan according to the uridine diphosphate glucuronosyltransferase (UGT) 1A1 genotype reflects the principle of individualized and precision medicine, and may improve the chemotherapy response and survival of CRC. Irinotecan 22-32 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 50-103 35356222-3 2022 Methods: To summarize the feasibility, efficacy and safety of high dose irinotecan in CRC patients with UGT1A1 wild-type or heterozygous alleles, PubMed, EMBASE, MEDLINE and the Cochrane Central Register of Controlled Trials online databases were searched from the date of creation to October 22, 2021. Irinotecan 72-82 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 104-110 35356222-5 2022 The results indicated that the maximum tolerated dose of irinotecan in CRC patients with UGT1A1 wild-type or heterozygous variant was significantly higher than the conventional recommended dose. Irinotecan 57-67 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 89-95 35356222-6 2022 Chemotherapy based on high dose irinotecan improved the clinical efficacy in mCRC patients with UGT1A1*28 wild-type and heterozygous variant, and the toxicity was tolerated, as reflected in most studies. Irinotecan 32-42 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 96-102 35356222-7 2022 Conclusions: We are optimistic about the application of high dose irinotecan for mCRC patients with UGT1A1*28 wild-type or heterozygous variant, which will provide a relatively clear direction for future research and certain norms for clinical practice. Irinotecan 66-76 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 100-106