PMID-sentid Pub_year Sent_text compound_name comp_offset prot_official_name organism prot_offset 31719901-12 2019 Conclusions: Ultrasound enhanced anti-tumor effect of temozolomide in glioblastoma cells via JNK and p38 pathways. temozolomide 54-66 mitogen-activated protein kinase 14 Mus musculus 101-104 31758290-7 2020 While TMZ treated cells did not show alteration in any of the Wnt ligands, PI3K inhibitor (LY294002) treatment repressed Akt activation and abolished the TMZ-mediated induction of Wnt/beta-catenin pathway. temozolomide 154-157 catenin beta 1 Homo sapiens 184-196 31671279-0 2020 Eukaryotic initiation factor 5B (eIF5B) regulates temozolomide-mediated apoptosis in brain tumor stem cells (BTSCs). temozolomide 50-62 eukaryotic translation initiation factor 5B Homo sapiens 0-31 31671279-0 2020 Eukaryotic initiation factor 5B (eIF5B) regulates temozolomide-mediated apoptosis in brain tumor stem cells (BTSCs). temozolomide 50-62 eukaryotic translation initiation factor 5B Homo sapiens 33-38 31671279-9 2020 We show that silencing eIF5B leads to increased TMZ sensitivity in two BTSC lines, BT25 and BT48. temozolomide 48-51 eukaryotic translation initiation factor 5B Homo sapiens 23-28 31671279-10 2020 Depletion of eIF5B decreases levels of anti-apoptotic proteins in BT48 and sensitizes these cells to TMZ-induced activation of caspase-3, cleavage of PARP, and apoptosis. temozolomide 101-104 eukaryotic translation initiation factor 5B Homo sapiens 13-18 31671279-10 2020 Depletion of eIF5B decreases levels of anti-apoptotic proteins in BT48 and sensitizes these cells to TMZ-induced activation of caspase-3, cleavage of PARP, and apoptosis. temozolomide 101-104 caspase 3 Homo sapiens 127-136 31671279-10 2020 Depletion of eIF5B decreases levels of anti-apoptotic proteins in BT48 and sensitizes these cells to TMZ-induced activation of caspase-3, cleavage of PARP, and apoptosis. temozolomide 101-104 poly(ADP-ribose) polymerase 1 Homo sapiens 150-154 31863352-9 2020 In cases of non-MGMT promoter methylation, systemic therapy with temozolomide is still recommended in many hospitals. temozolomide 65-77 O-6-methylguanine-DNA methyltransferase Homo sapiens 16-20 31865606-0 2020 IKBKE enhances TMZ-chemoresistance through upregulation of MGMT expression in glioblastoma. temozolomide 15-18 inhibitor of nuclear factor kappa B kinase subunit epsilon Homo sapiens 0-5 31865606-0 2020 IKBKE enhances TMZ-chemoresistance through upregulation of MGMT expression in glioblastoma. temozolomide 15-18 O-6-methylguanine-DNA methyltransferase Homo sapiens 59-63 31865606-5 2020 Our study aimed to investigate the mechanism of IKBKE enhancing the resistance of glioma cells to temozolomide. temozolomide 98-110 inhibitor of nuclear factor kappa B kinase subunit epsilon Homo sapiens 48-53 31865606-11 2020 RESULTS: In this study, we demonstrated that IKBKE enhances the resistance of glioblastoma cells to temozolomide (TMZ) by activating the AKT/NF-kappaB signaling pathway to upregulate the expression of the DNA repair enzyme o6-methylguanine-dna methyltransferase (MGMT). temozolomide 100-112 inhibitor of nuclear factor kappa B kinase subunit epsilon Homo sapiens 45-50 31865606-11 2020 RESULTS: In this study, we demonstrated that IKBKE enhances the resistance of glioblastoma cells to temozolomide (TMZ) by activating the AKT/NF-kappaB signaling pathway to upregulate the expression of the DNA repair enzyme o6-methylguanine-dna methyltransferase (MGMT). temozolomide 100-112 AKT serine/threonine kinase 1 Homo sapiens 137-140 31865606-11 2020 RESULTS: In this study, we demonstrated that IKBKE enhances the resistance of glioblastoma cells to temozolomide (TMZ) by activating the AKT/NF-kappaB signaling pathway to upregulate the expression of the DNA repair enzyme o6-methylguanine-dna methyltransferase (MGMT). temozolomide 114-117 inhibitor of nuclear factor kappa B kinase subunit epsilon Homo sapiens 45-50 31865606-11 2020 RESULTS: In this study, we demonstrated that IKBKE enhances the resistance of glioblastoma cells to temozolomide (TMZ) by activating the AKT/NF-kappaB signaling pathway to upregulate the expression of the DNA repair enzyme o6-methylguanine-dna methyltransferase (MGMT). temozolomide 114-117 AKT serine/threonine kinase 1 Homo sapiens 137-140 31865606-11 2020 RESULTS: In this study, we demonstrated that IKBKE enhances the resistance of glioblastoma cells to temozolomide (TMZ) by activating the AKT/NF-kappaB signaling pathway to upregulate the expression of the DNA repair enzyme o6-methylguanine-dna methyltransferase (MGMT). temozolomide 114-117 O-6-methylguanine-DNA methyltransferase Homo sapiens 223-261 31865606-11 2020 RESULTS: In this study, we demonstrated that IKBKE enhances the resistance of glioblastoma cells to temozolomide (TMZ) by activating the AKT/NF-kappaB signaling pathway to upregulate the expression of the DNA repair enzyme o6-methylguanine-dna methyltransferase (MGMT). temozolomide 114-117 O-6-methylguanine-DNA methyltransferase Homo sapiens 263-267 31865606-12 2020 In glioblastoma cells, IKBKE knockdown enhances apoptosis and suppresses cell proliferation, clone formation, and tumor development in vivo induced by TMZ. temozolomide 151-154 inhibitor of nuclear factor kappa B kinase subunit epsilon Homo sapiens 23-28 31865606-13 2020 However, overexpression of IKBKE reduces the effects of TMZ. temozolomide 56-59 inhibitor of nuclear factor kappa B kinase subunit epsilon Homo sapiens 27-32 31865606-14 2020 CONCLUSION: Our studies suggest that inhibition of IKBKE can enhance the therapeutic effect of TMZ on GBM in vitro and in vivo, providing new research directions and therapeutic targets for the treatment of GBM. temozolomide 95-98 inhibitor of nuclear factor kappa B kinase subunit epsilon Homo sapiens 51-56 31916238-0 2020 A Key Role of DNA Damage-Inducible Transcript 4 (DDIT4) Connects Autophagy and GLUT3-Mediated Stemness To Desensitize Temozolomide Efficacy in Glioblastomas. temozolomide 118-130 DNA damage inducible transcript 4 Homo sapiens 14-47 31916238-0 2020 A Key Role of DNA Damage-Inducible Transcript 4 (DDIT4) Connects Autophagy and GLUT3-Mediated Stemness To Desensitize Temozolomide Efficacy in Glioblastomas. temozolomide 118-130 DNA damage inducible transcript 4 Homo sapiens 49-54 31916238-0 2020 A Key Role of DNA Damage-Inducible Transcript 4 (DDIT4) Connects Autophagy and GLUT3-Mediated Stemness To Desensitize Temozolomide Efficacy in Glioblastomas. temozolomide 118-130 solute carrier family 2 member 3 Homo sapiens 79-84 31916238-3 2020 Herein, we investigated DDIT4 signaling in GBM and temozolomide (TMZ) drug resistance. temozolomide 65-68 DNA damage inducible transcript 4 Homo sapiens 24-29 31916238-4 2020 We identified that TMZ induced DDIT4 upregulation, leading to desensitization against TMZ cytotoxicity in GBM cells. temozolomide 19-22 DNA damage inducible transcript 4 Homo sapiens 31-36 31916238-4 2020 We identified that TMZ induced DDIT4 upregulation, leading to desensitization against TMZ cytotoxicity in GBM cells. temozolomide 86-89 DNA damage inducible transcript 4 Homo sapiens 31-36 31916238-6 2020 Furthermore, patients with lower DDIT4 levels, especially O-6-methylguanine-DNA methyltransferase (MGMT)-methylated patients, exhibited better TMZ therapeutic efficacy. temozolomide 143-146 DNA damage inducible transcript 4 Homo sapiens 33-38 31916238-6 2020 Furthermore, patients with lower DDIT4 levels, especially O-6-methylguanine-DNA methyltransferase (MGMT)-methylated patients, exhibited better TMZ therapeutic efficacy. temozolomide 143-146 O-6-methylguanine-DNA methyltransferase Homo sapiens 58-97 31916238-6 2020 Furthermore, patients with lower DDIT4 levels, especially O-6-methylguanine-DNA methyltransferase (MGMT)-methylated patients, exhibited better TMZ therapeutic efficacy. temozolomide 143-146 O-6-methylguanine-DNA methyltransferase Homo sapiens 99-103 31916238-8 2020 Among these 5 genes, only GLUT3 was upregulated in both TMZ-treated and DDIT4-overexpressing cells. temozolomide 56-59 solute carrier family 2 member 3 Homo sapiens 26-31 31916238-9 2020 DDIT4-mediated GLUT3 expression was also identified, and its expression decreased TMZ"s cytotoxicity. temozolomide 82-85 DNA damage inducible transcript 4 Homo sapiens 0-5 31916238-9 2020 DDIT4-mediated GLUT3 expression was also identified, and its expression decreased TMZ"s cytotoxicity. temozolomide 82-85 solute carrier family 2 member 3 Homo sapiens 15-20 31916238-13 2020 Finally, we identified TMZ-upregulated activating transcription factor 4 (ATF4) as an upstream regulator of DDIT4-mediated GLUT3/stemness signaling and autophagy. temozolomide 23-26 activating transcription factor 4 Homo sapiens 39-72 31916238-13 2020 Finally, we identified TMZ-upregulated activating transcription factor 4 (ATF4) as an upstream regulator of DDIT4-mediated GLUT3/stemness signaling and autophagy. temozolomide 23-26 activating transcription factor 4 Homo sapiens 74-78 31916238-13 2020 Finally, we identified TMZ-upregulated activating transcription factor 4 (ATF4) as an upstream regulator of DDIT4-mediated GLUT3/stemness signaling and autophagy. temozolomide 23-26 DNA damage inducible transcript 4 Homo sapiens 108-113 31916238-13 2020 Finally, we identified TMZ-upregulated activating transcription factor 4 (ATF4) as an upstream regulator of DDIT4-mediated GLUT3/stemness signaling and autophagy. temozolomide 23-26 solute carrier family 2 member 3 Homo sapiens 123-128 31916238-14 2020 Consequently, ATF4/DDIT4 signaling was connected to both autophagy and GLUT3-regulated stemness, which are involved in TMZ drug resistance and the poor prognoses of GBM patients. temozolomide 119-122 activating transcription factor 4 Homo sapiens 14-18 31916238-14 2020 Consequently, ATF4/DDIT4 signaling was connected to both autophagy and GLUT3-regulated stemness, which are involved in TMZ drug resistance and the poor prognoses of GBM patients. temozolomide 119-122 DNA damage inducible transcript 4 Homo sapiens 19-24 31916238-14 2020 Consequently, ATF4/DDIT4 signaling was connected to both autophagy and GLUT3-regulated stemness, which are involved in TMZ drug resistance and the poor prognoses of GBM patients. temozolomide 119-122 solute carrier family 2 member 3 Homo sapiens 71-76 31578804-0 2020 Receptor tyrosine kinase AXL is correlated with poor prognosis and induces temozolomide resistance in glioblastoma. temozolomide 75-87 AXL receptor tyrosine kinase Homo sapiens 25-28 31578804-1 2020 AIMS: To investigate the functions and underlying mechanisms of AXL receptor tyrosine kinase (AXL) in tumor proliferation and chemoresistance to temozolomide (TMZ) in glioblastoma (GBM). temozolomide 145-157 AXL receptor tyrosine kinase Homo sapiens 64-67 31578804-1 2020 AIMS: To investigate the functions and underlying mechanisms of AXL receptor tyrosine kinase (AXL) in tumor proliferation and chemoresistance to temozolomide (TMZ) in glioblastoma (GBM). temozolomide 145-157 AXL receptor tyrosine kinase Homo sapiens 94-97 31578804-1 2020 AIMS: To investigate the functions and underlying mechanisms of AXL receptor tyrosine kinase (AXL) in tumor proliferation and chemoresistance to temozolomide (TMZ) in glioblastoma (GBM). temozolomide 159-162 AXL receptor tyrosine kinase Homo sapiens 64-67 31578804-1 2020 AIMS: To investigate the functions and underlying mechanisms of AXL receptor tyrosine kinase (AXL) in tumor proliferation and chemoresistance to temozolomide (TMZ) in glioblastoma (GBM). temozolomide 159-162 AXL receptor tyrosine kinase Homo sapiens 94-97 31578804-2 2020 METHODS: With a kinome-wide bioinformatics analysis, AXL was found to be an essential kinase candidate in TMZ chemoresistance promotion. temozolomide 106-109 AXL receptor tyrosine kinase Homo sapiens 53-56 31578804-3 2020 Additionally, the biological functions of AXL in oncogenesis and TMZ resistance were clarified by using qRT-PCR, Western blotting, and in vivo intracranial GBM xenograft models followed the induction of TMZ resistance in U87 or U251 cells. temozolomide 203-206 AXL receptor tyrosine kinase Homo sapiens 42-45 31578804-5 2020 Finally, the Chou-Talalay model was performed to confirm the synergistic effect of AXL inhibitor TP-0903 with TMZ. temozolomide 110-113 AXL receptor tyrosine kinase Homo sapiens 83-86 31578804-7 2020 Moreover, AXL knockdown reduced tumorigenesis and TMZ resistance in vitro and in vivo; however, exogenous AXL overexpression induced TMZ resistance in GBM. temozolomide 50-53 AXL receptor tyrosine kinase Homo sapiens 10-13 31578804-7 2020 Moreover, AXL knockdown reduced tumorigenesis and TMZ resistance in vitro and in vivo; however, exogenous AXL overexpression induced TMZ resistance in GBM. temozolomide 133-136 AXL receptor tyrosine kinase Homo sapiens 106-109 31578804-8 2020 Lastly, a specific AXL inhibitor, TP-0903, dramatically decreased tumor growth and increased sensitivity to TMZ via a synergistic effect. temozolomide 108-111 AXL receptor tyrosine kinase Homo sapiens 19-22 31578804-9 2020 CONCLUSION: AXL contributed to chemoresistance to TMZ in GBM and could be used as a novel prognostic biomarker and therapeutic target for GBM. temozolomide 50-53 AXL receptor tyrosine kinase Homo sapiens 12-15 31853548-6 2020 Studies in animal models documented that FUS enhanced the delivery of numerous chemotherapeutic and investigational agents across the BBB and into brain tumors, including temozolomide, bevacizumab, 1,3-bis (2-chloroethyl)-1-nitrosourea, doxorubicin, viral vectors, and cells. temozolomide 171-183 FUS RNA binding protein Homo sapiens 41-44 31758290-0 2020 Temozolomide induces activation of Wnt/beta-catenin signaling in glioma cells via PI3K/Akt pathway: implications in glioma therapy. temozolomide 0-12 catenin beta 1 Homo sapiens 39-51 31758290-0 2020 Temozolomide induces activation of Wnt/beta-catenin signaling in glioma cells via PI3K/Akt pathway: implications in glioma therapy. temozolomide 0-12 AKT serine/threonine kinase 1 Homo sapiens 87-90 31758290-5 2020 Gene Set Enrichment Analysis (GSEA) indicated a significant enrichment of Wnt/beta-catenin signaling besides many other pathways in TMZ treated cells. temozolomide 132-135 catenin beta 1 Homo sapiens 78-90 31758290-6 2020 Further, we demonstrate that TMZ treatment increased the activity from TOPflash reporter, (a Wnt responsive reporter), enhanced the levels of pGSK-3beta (S9) and reduced the levels of p-beta-catenin (S33/37/T41) with a concomitant increase in transcript and protein levels of Wnt targets in a concentration and time-dependent manner. temozolomide 29-32 catenin beta 1 Homo sapiens 186-198 31758290-8 2020 In addition, we show that Wnt/beta-catenin signaling activation by TMZ is independent of ATM/Chk2 pathway. temozolomide 67-70 catenin beta 1 Homo sapiens 30-42 31758290-9 2020 Further, we also demonstrate the activation of mTOR pathway after TMZ treatment. temozolomide 66-69 mechanistic target of rapamycin kinase Homo sapiens 47-51 31758290-10 2020 Thus, our results demonstrate that activation of Wnt/beta-catenin pathway involves an ATM/Chk2- independent PI3K/Akt/GSK-3 cascade in TMZ treated cells and further provides mechanistic basis for the chemoresistance of glioma to TMZ. temozolomide 134-137 catenin beta 1 Homo sapiens 53-65 31758290-10 2020 Thus, our results demonstrate that activation of Wnt/beta-catenin pathway involves an ATM/Chk2- independent PI3K/Akt/GSK-3 cascade in TMZ treated cells and further provides mechanistic basis for the chemoresistance of glioma to TMZ. temozolomide 134-137 ATM serine/threonine kinase Homo sapiens 86-89 31758290-10 2020 Thus, our results demonstrate that activation of Wnt/beta-catenin pathway involves an ATM/Chk2- independent PI3K/Akt/GSK-3 cascade in TMZ treated cells and further provides mechanistic basis for the chemoresistance of glioma to TMZ. temozolomide 134-137 checkpoint kinase 2 Homo sapiens 90-94 31758290-10 2020 Thus, our results demonstrate that activation of Wnt/beta-catenin pathway involves an ATM/Chk2- independent PI3K/Akt/GSK-3 cascade in TMZ treated cells and further provides mechanistic basis for the chemoresistance of glioma to TMZ. temozolomide 134-137 AKT serine/threonine kinase 1 Homo sapiens 113-116 31758290-10 2020 Thus, our results demonstrate that activation of Wnt/beta-catenin pathway involves an ATM/Chk2- independent PI3K/Akt/GSK-3 cascade in TMZ treated cells and further provides mechanistic basis for the chemoresistance of glioma to TMZ. temozolomide 228-231 catenin beta 1 Homo sapiens 53-65 31758290-10 2020 Thus, our results demonstrate that activation of Wnt/beta-catenin pathway involves an ATM/Chk2- independent PI3K/Akt/GSK-3 cascade in TMZ treated cells and further provides mechanistic basis for the chemoresistance of glioma to TMZ. temozolomide 228-231 ATM serine/threonine kinase Homo sapiens 86-89 31987922-7 2020 Moreover, Nano-DOX also suppressed stimulated activation of STAT3 and IL-6 induced by temozolomide, a first-line anti-GBM chemotherapy, resistance to which critically involves STAT3 activation. temozolomide 86-98 signal transducer and activator of transcription 3 Homo sapiens 60-65 32045717-5 2020 TMZ inhibited mtDNA replication and transcription of mitochondrial genes (ND1 and Cyt b) in NSC by 24 h, whereas the effect of TMZ on neuronal mtDNA transcription was less pronounced. temozolomide 0-3 mitochondrially encoded NADH dehydrogenase 1 Homo sapiens 74-77 32045717-5 2020 TMZ inhibited mtDNA replication and transcription of mitochondrial genes (ND1 and Cyt b) in NSC by 24 h, whereas the effect of TMZ on neuronal mtDNA transcription was less pronounced. temozolomide 0-3 mitochondrially encoded cytochrome b Homo sapiens 82-87 32045717-7 2020 Acute TMZ exposure (4 h) caused a rapid reduction in dendritic branching and loss of postsynaptic density-95 (PSD95) puncta on dendrites. temozolomide 6-9 discs large MAGUK scaffold protein 4 Homo sapiens 85-108 32045717-7 2020 Acute TMZ exposure (4 h) caused a rapid reduction in dendritic branching and loss of postsynaptic density-95 (PSD95) puncta on dendrites. temozolomide 6-9 discs large MAGUK scaffold protein 4 Homo sapiens 110-115 32032585-14 2020 Organoids with reduced levels of MGMT and CHFR mRNAs were sensitive to temozolomide and taxane drugs. temozolomide 71-83 O-6-methylguanine-DNA methyltransferase Homo sapiens 33-37 32032585-14 2020 Organoids with reduced levels of MGMT and CHFR mRNAs were sensitive to temozolomide and taxane drugs. temozolomide 71-83 checkpoint with forkhead and ring finger domains Homo sapiens 42-46 31987922-7 2020 Moreover, Nano-DOX also suppressed stimulated activation of STAT3 and IL-6 induced by temozolomide, a first-line anti-GBM chemotherapy, resistance to which critically involves STAT3 activation. temozolomide 86-98 interleukin 6 Homo sapiens 70-74 31987922-7 2020 Moreover, Nano-DOX also suppressed stimulated activation of STAT3 and IL-6 induced by temozolomide, a first-line anti-GBM chemotherapy, resistance to which critically involves STAT3 activation. temozolomide 86-98 signal transducer and activator of transcription 3 Homo sapiens 176-181 31833081-3 2020 The aims of this study were to comprehend the effect of TMZ on nucleus and the underlying mechanism of acquired TMZ resistance in MGMT-deficient GBM. temozolomide 56-59 O-6-methylguanine-DNA methyltransferase Homo sapiens 130-134 31900275-7 2020 RESULTS: While having no detectable effect in MSH6 wild-type GBMs, PARPi selectively restored TMZ sensitivity in MSH6-deficient GBM cells. temozolomide 94-97 mutS homolog 6 Homo sapiens 113-117 31900275-8 2020 This genotype-specific restoration of activity translated in vivo, where combination treatment of veliparib and TMZ showed potent suppression of tumor growth of MSH6-inactivated orthotopic xenografts, compared with TMZ monotherapy. temozolomide 112-115 mutS homolog 6 Homo sapiens 161-165 31900275-11 2020 CONCLUSIONS: PARPi restoration of TMZ chemosensitivity in MSH6-inactivated glioma represents a promising strategy to overcome acquired chemoresistance caused by MMR deficiency. temozolomide 34-37 mutS homolog 6 Homo sapiens 58-62 31613002-0 2020 Role of Sonic hedgehog signaling in cell cycle, oxidative stress, and autophagy of temozolomide resistant glioblastoma. temozolomide 83-95 sonic hedgehog signaling molecule Homo sapiens 8-22 31613002-2 2020 The Sonic hedgehog (SHH) pathway is involved with GBM tumorigenesis and TMZ chemoresistance. temozolomide 72-75 sonic hedgehog signaling molecule Homo sapiens 4-18 31613002-2 2020 The Sonic hedgehog (SHH) pathway is involved with GBM tumorigenesis and TMZ chemoresistance. temozolomide 72-75 sonic hedgehog signaling molecule Homo sapiens 20-23 31613002-3 2020 The role of SHH pathway inhibition in the potentiation of TMZ"s effects using T98G, U251, and GBM11 cell lines is investigated herein. temozolomide 58-61 sonic hedgehog signaling molecule Homo sapiens 12-15 31613002-9 2020 The selective inhibition of the SHH pathway potentiated the cytotoxic effect of TMZ, thus becoming a promising in vitro strategy for GBM treatment. temozolomide 80-83 sonic hedgehog signaling molecule Homo sapiens 32-35 31750728-0 2020 NCK1-AS1 Increases Drug Resistance of Glioma Cells to Temozolomide by Modulating miR-137/TRIM24. temozolomide 54-66 NCK adaptor protein 1 Homo sapiens 0-4 31750728-0 2020 NCK1-AS1 Increases Drug Resistance of Glioma Cells to Temozolomide by Modulating miR-137/TRIM24. temozolomide 54-66 prostaglandin D2 receptor Homo sapiens 5-8 32036239-0 2020 Temozolomide for patients with wild-type isocitrate dehydrogenase (IDH) 1 glioblastoma using propensity score matching. temozolomide 0-12 isocitrate dehydrogenase (NADP(+)) 1 Homo sapiens 67-73 32036239-1 2020 OBJECTIVE: The isocitrate dehydrogenase (IDH) 1 wild-type glioblastoma (GBM) is a major population of GBM that should be of concern in terms of the efficacy of using Temozolomide (TMZ) in adjuvant treatment. temozolomide 166-178 isocitrate dehydrogenase (NADP(+)) 1 Homo sapiens 15-47 32036239-1 2020 OBJECTIVE: The isocitrate dehydrogenase (IDH) 1 wild-type glioblastoma (GBM) is a major population of GBM that should be of concern in terms of the efficacy of using Temozolomide (TMZ) in adjuvant treatment. temozolomide 180-183 isocitrate dehydrogenase (NADP(+)) 1 Homo sapiens 15-47 32036239-13 2020 CONCLUSIONS: The present study revealed that the effect of TMZ with RT in IDH1 wild-type GBM patients had advantages in terms of survival by using multivariable analysis and PS approaches. temozolomide 59-62 isocitrate dehydrogenase (NADP(+)) 1 Homo sapiens 74-78 31750728-0 2020 NCK1-AS1 Increases Drug Resistance of Glioma Cells to Temozolomide by Modulating miR-137/TRIM24. temozolomide 54-66 microRNA 137 Homo sapiens 81-88 31750728-0 2020 NCK1-AS1 Increases Drug Resistance of Glioma Cells to Temozolomide by Modulating miR-137/TRIM24. temozolomide 54-66 tripartite motif containing 24 Homo sapiens 89-95 31750728-2 2020 The purpose of this study was to investigate the effect and molecular mechanism of long noncoding RNA (lncRNA) NCK1-AS1 on the drug resistance of temozolomide (TMZ) in glioma cells. temozolomide 146-158 NCK adaptor protein 1 Homo sapiens 111-115 31750728-2 2020 The purpose of this study was to investigate the effect and molecular mechanism of long noncoding RNA (lncRNA) NCK1-AS1 on the drug resistance of temozolomide (TMZ) in glioma cells. temozolomide 146-158 prostaglandin D2 receptor Homo sapiens 116-119 31750728-2 2020 The purpose of this study was to investigate the effect and molecular mechanism of long noncoding RNA (lncRNA) NCK1-AS1 on the drug resistance of temozolomide (TMZ) in glioma cells. temozolomide 160-163 NCK adaptor protein 1 Homo sapiens 111-115 31750728-2 2020 The purpose of this study was to investigate the effect and molecular mechanism of long noncoding RNA (lncRNA) NCK1-AS1 on the drug resistance of temozolomide (TMZ) in glioma cells. temozolomide 160-163 prostaglandin D2 receptor Homo sapiens 116-119 31750728-8 2020 Results: NCK1-AS1 expression was increased in regular and recurrent glioma tissues and TMZ-resistant cells. temozolomide 87-90 NCK adaptor protein 1 Homo sapiens 9-13 31750728-8 2020 Results: NCK1-AS1 expression was increased in regular and recurrent glioma tissues and TMZ-resistant cells. temozolomide 87-90 prostaglandin D2 receptor Homo sapiens 14-17 31750728-13 2020 In addition, the decreases of cell viability and IC50 induced by NCK1-AS1 knockdown were reversed after adding TRIM24 in U251/TMZ and A172/TMZ cells. temozolomide 126-129 NCK adaptor protein 1 Homo sapiens 65-69 31750728-13 2020 In addition, the decreases of cell viability and IC50 induced by NCK1-AS1 knockdown were reversed after adding TRIM24 in U251/TMZ and A172/TMZ cells. temozolomide 126-129 prostaglandin D2 receptor Homo sapiens 70-73 31750728-13 2020 In addition, the decreases of cell viability and IC50 induced by NCK1-AS1 knockdown were reversed after adding TRIM24 in U251/TMZ and A172/TMZ cells. temozolomide 126-129 tripartite motif containing 24 Homo sapiens 111-117 31750728-13 2020 In addition, the decreases of cell viability and IC50 induced by NCK1-AS1 knockdown were reversed after adding TRIM24 in U251/TMZ and A172/TMZ cells. temozolomide 139-142 NCK adaptor protein 1 Homo sapiens 65-69 31750728-13 2020 In addition, the decreases of cell viability and IC50 induced by NCK1-AS1 knockdown were reversed after adding TRIM24 in U251/TMZ and A172/TMZ cells. temozolomide 139-142 prostaglandin D2 receptor Homo sapiens 70-73 31750728-13 2020 In addition, the decreases of cell viability and IC50 induced by NCK1-AS1 knockdown were reversed after adding TRIM24 in U251/TMZ and A172/TMZ cells. temozolomide 139-142 tripartite motif containing 24 Homo sapiens 111-117 31750728-14 2020 Conclusion: NCK1-AS1 could increase drug resistance of glioma cells to TMZ by modulating miR-137/TRIM24 pathway. temozolomide 71-74 NCK adaptor protein 1 Homo sapiens 12-16 31750728-14 2020 Conclusion: NCK1-AS1 could increase drug resistance of glioma cells to TMZ by modulating miR-137/TRIM24 pathway. temozolomide 71-74 prostaglandin D2 receptor Homo sapiens 17-20 31750728-14 2020 Conclusion: NCK1-AS1 could increase drug resistance of glioma cells to TMZ by modulating miR-137/TRIM24 pathway. temozolomide 71-74 microRNA 137 Homo sapiens 89-96 31750728-14 2020 Conclusion: NCK1-AS1 could increase drug resistance of glioma cells to TMZ by modulating miR-137/TRIM24 pathway. temozolomide 71-74 tripartite motif containing 24 Homo sapiens 97-103 31833081-3 2020 The aims of this study were to comprehend the effect of TMZ on nucleus and the underlying mechanism of acquired TMZ resistance in MGMT-deficient GBM. temozolomide 112-115 O-6-methylguanine-DNA methyltransferase Homo sapiens 130-134 31833081-4 2020 We show the changes of nuclear proteome in the MGMT-deficient GBM U87 cells treated with TMZ for 1 week. temozolomide 89-92 O-6-methylguanine-DNA methyltransferase Homo sapiens 47-51 32070670-10 2020 When the patients were divided into pre and post temozolomide era, the association of MGMT promoter methylation status with PFS was only noted in the patients of temozolomide era (pre, p = 0.432; post, p = 0.015). temozolomide 49-61 O-6-methylguanine-DNA methyltransferase Homo sapiens 86-90 32070670-10 2020 When the patients were divided into pre and post temozolomide era, the association of MGMT promoter methylation status with PFS was only noted in the patients of temozolomide era (pre, p = 0.432; post, p = 0.015). temozolomide 162-174 O-6-methylguanine-DNA methyltransferase Homo sapiens 86-90 31806377-5 2020 Interestingly, we observed that the addition of four different autophagy inducers, rapamycin (RAP), CCI779, ABT737 and temozolomide (TMZ), induced the differences of DR5 expression and cell apoptosis after irradiation. temozolomide 119-131 TNF receptor superfamily member 10b Homo sapiens 166-169 31756732-9 2020 MSI1 was found to promote drug resistance to the combined treatment with TMZ and VPA. temozolomide 73-76 musashi RNA binding protein 1 Homo sapiens 0-4 31806377-5 2020 Interestingly, we observed that the addition of four different autophagy inducers, rapamycin (RAP), CCI779, ABT737 and temozolomide (TMZ), induced the differences of DR5 expression and cell apoptosis after irradiation. temozolomide 133-136 TNF receptor superfamily member 10b Homo sapiens 166-169 31806377-6 2020 Unlike RAP and CCI779, ABT737 and TMZ were able to increase DR5 expression and further induce cell death. temozolomide 34-37 TNF receptor superfamily member 10b Homo sapiens 60-63 32020475-7 2020 A Cox proportional hazards model identified extent of resection and MGMT methylation status as significant prognostic factors in the TMZ era; however, these factors were not significant in the TMZ-BEV era. temozolomide 133-136 O-6-methylguanine-DNA methyltransferase Homo sapiens 68-72 32028734-1 2020 13a-(S)-3-pivaloyloxyl-6,7-dimethoxyphenanthro(9,10-b)-indolizidine (CAT3) is a novel oral anti-glioma pro-drug with a potent anti-tumor effect against temozolomide-resistant glioma in vivo. temozolomide 152-164 solute carrier family 7 member 3 Rattus norvegicus 69-73 31841318-8 2020 The DNA repair protein MGMT (O6-methylguanine DNA methyltransferase) protects against environmental mutagenesis by DNA methylating agents and, by removing m6G, limits the therapeutic potential of temozolomide in cancer therapy. temozolomide 196-208 O-6-methylguanine-DNA methyltransferase Mus musculus 23-27 31841318-8 2020 The DNA repair protein MGMT (O6-methylguanine DNA methyltransferase) protects against environmental mutagenesis by DNA methylating agents and, by removing m6G, limits the therapeutic potential of temozolomide in cancer therapy. temozolomide 196-208 O-6-methylguanine-DNA methyltransferase Mus musculus 29-67 32075134-6 2020 Visualization of active MGMT levels reveals discrete active and inactive MGMT populations with biphasic kinetics for MGMT inactivation in response to TMZ-induced DNA damage. temozolomide 150-153 O-6-methylguanine-DNA methyltransferase Homo sapiens 24-28 32075134-6 2020 Visualization of active MGMT levels reveals discrete active and inactive MGMT populations with biphasic kinetics for MGMT inactivation in response to TMZ-induced DNA damage. temozolomide 150-153 O-6-methylguanine-DNA methyltransferase Homo sapiens 73-77 32075134-6 2020 Visualization of active MGMT levels reveals discrete active and inactive MGMT populations with biphasic kinetics for MGMT inactivation in response to TMZ-induced DNA damage. temozolomide 150-153 O-6-methylguanine-DNA methyltransferase Homo sapiens 73-77 32020475-8 2020 In subgroup analyses, patients with MGMT methylation had improved OS after TMZ introduction (pre-TMZ vs. TMZ, 18.5 vs. 28.1 months; P = 0.13), and those without MGMT methylation had significantly increased OS after BEV approval (TMZ vs. TMZ-BEV, 12.2 vs. 16.7 months; P = 0.04). temozolomide 97-100 O-6-methylguanine-DNA methyltransferase Homo sapiens 36-40 32020475-8 2020 In subgroup analyses, patients with MGMT methylation had improved OS after TMZ introduction (pre-TMZ vs. TMZ, 18.5 vs. 28.1 months; P = 0.13), and those without MGMT methylation had significantly increased OS after BEV approval (TMZ vs. TMZ-BEV, 12.2 vs. 16.7 months; P = 0.04). temozolomide 97-100 O-6-methylguanine-DNA methyltransferase Homo sapiens 36-40 32020475-8 2020 In subgroup analyses, patients with MGMT methylation had improved OS after TMZ introduction (pre-TMZ vs. TMZ, 18.5 vs. 28.1 months; P = 0.13), and those without MGMT methylation had significantly increased OS after BEV approval (TMZ vs. TMZ-BEV, 12.2 vs. 16.7 months; P = 0.04). temozolomide 75-78 O-6-methylguanine-DNA methyltransferase Homo sapiens 36-40 32020475-8 2020 In subgroup analyses, patients with MGMT methylation had improved OS after TMZ introduction (pre-TMZ vs. TMZ, 18.5 vs. 28.1 months; P = 0.13), and those without MGMT methylation had significantly increased OS after BEV approval (TMZ vs. TMZ-BEV, 12.2 vs. 16.7 months; P = 0.04). temozolomide 97-100 O-6-methylguanine-DNA methyltransferase Homo sapiens 36-40 32020475-8 2020 In subgroup analyses, patients with MGMT methylation had improved OS after TMZ introduction (pre-TMZ vs. TMZ, 18.5 vs. 28.1 months; P = 0.13), and those without MGMT methylation had significantly increased OS after BEV approval (TMZ vs. TMZ-BEV, 12.2 vs. 16.7 months; P = 0.04). temozolomide 97-100 O-6-methylguanine-DNA methyltransferase Homo sapiens 36-40 31896509-0 2020 AR ubiquitination induced by the curcumin analog suppresses growth of temozolomide-resistant glioblastoma through disrupting GPX4-Mediated redox homeostasis. temozolomide 70-82 androgen receptor Mus musculus 0-2 31653969-12 2020 In glioblastoma, the TRPV2 is part of an interactome-based signature complex, which is negatively associated with survival, and it is expressed in high risk of recurrence and temozolomide-resistant patients. temozolomide 175-187 transient receptor potential cation channel subfamily V member 2 Homo sapiens 21-26 31896509-0 2020 AR ubiquitination induced by the curcumin analog suppresses growth of temozolomide-resistant glioblastoma through disrupting GPX4-Mediated redox homeostasis. temozolomide 70-82 glutathione peroxidase 4 Mus musculus 125-129 31896509-12 2020 Taken together, AR potentiates TMZ resistance for glioblastoma, and ALZ003-mediated AR ubiquitination might open a new insight into therapeutic strategy for TMZ resistant glioblastoma. temozolomide 157-160 androgen receptor Mus musculus 84-86 31896509-5 2020 In this study, we found that AR expression is significantly correlated with poor prognosis, and AR obviously induced the resistance to temozolomide (TMZ) treatment. temozolomide 135-147 androgen receptor Mus musculus 96-98 31896509-5 2020 In this study, we found that AR expression is significantly correlated with poor prognosis, and AR obviously induced the resistance to temozolomide (TMZ) treatment. temozolomide 149-152 androgen receptor Mus musculus 96-98 31896509-12 2020 Taken together, AR potentiates TMZ resistance for glioblastoma, and ALZ003-mediated AR ubiquitination might open a new insight into therapeutic strategy for TMZ resistant glioblastoma. temozolomide 31-34 androgen receptor Mus musculus 16-18 32003751-9 2020 Moreover, IL-33 prevented temozolomide induced tumor apoptosis. temozolomide 26-38 interleukin 33 Homo sapiens 10-15 32001707-0 2020 Dual functionalized brain-targeting nanoinhibitors restrain temozolomide-resistant glioma via attenuating EGFR and MET signaling pathways. temozolomide 60-72 epidermal growth factor receptor Homo sapiens 106-110 32001707-2 2020 In this study, the crosstalk activation of epidermal growth factor receptor (EGFR) and mesenchymal-epithelial transition factor (MET) signaling pathways is demonstrated to contribute to temozolomide (TMZ) resistance, resulting in an unfavorable prognosis for patients with glioblastoma. temozolomide 186-198 epidermal growth factor receptor Homo sapiens 43-75 32001707-2 2020 In this study, the crosstalk activation of epidermal growth factor receptor (EGFR) and mesenchymal-epithelial transition factor (MET) signaling pathways is demonstrated to contribute to temozolomide (TMZ) resistance, resulting in an unfavorable prognosis for patients with glioblastoma. temozolomide 186-198 epidermal growth factor receptor Homo sapiens 77-81 32001707-2 2020 In this study, the crosstalk activation of epidermal growth factor receptor (EGFR) and mesenchymal-epithelial transition factor (MET) signaling pathways is demonstrated to contribute to temozolomide (TMZ) resistance, resulting in an unfavorable prognosis for patients with glioblastoma. temozolomide 200-203 epidermal growth factor receptor Homo sapiens 43-75 32001707-2 2020 In this study, the crosstalk activation of epidermal growth factor receptor (EGFR) and mesenchymal-epithelial transition factor (MET) signaling pathways is demonstrated to contribute to temozolomide (TMZ) resistance, resulting in an unfavorable prognosis for patients with glioblastoma. temozolomide 200-203 epidermal growth factor receptor Homo sapiens 77-81 32003751-0 2020 The Interleukin-33/ST2 axis promotes glioma mesenchymal transition, stemness and TMZ resistance via JNK activation. temozolomide 81-84 interleukin 33 Homo sapiens 4-18 32003751-10 2020 Anti-ST2 or knockdown IL-33 increased the sensitivity of tumor to temozolomide. temozolomide 66-78 ST2 Homo sapiens 5-8 32003751-0 2020 The Interleukin-33/ST2 axis promotes glioma mesenchymal transition, stemness and TMZ resistance via JNK activation. temozolomide 81-84 ST2 Homo sapiens 19-22 32001707-4 2020 In the presence of BIP-MPC-NP, DNA damage repair is attenuated and TMZ sensitivity is enhanced via the down-regulation of E2F1 mediated by TTP in TMZ resistant glioma. temozolomide 67-70 growth differentiation factor 10 Homo sapiens 19-22 32001707-4 2020 In the presence of BIP-MPC-NP, DNA damage repair is attenuated and TMZ sensitivity is enhanced via the down-regulation of E2F1 mediated by TTP in TMZ resistant glioma. temozolomide 67-70 E2F transcription factor 1 Homo sapiens 122-126 32001707-4 2020 In the presence of BIP-MPC-NP, DNA damage repair is attenuated and TMZ sensitivity is enhanced via the down-regulation of E2F1 mediated by TTP in TMZ resistant glioma. temozolomide 146-149 growth differentiation factor 10 Homo sapiens 19-22 32003751-10 2020 Anti-ST2 or knockdown IL-33 increased the sensitivity of tumor to temozolomide. temozolomide 66-78 interleukin 33 Homo sapiens 22-27 32001707-4 2020 In the presence of BIP-MPC-NP, DNA damage repair is attenuated and TMZ sensitivity is enhanced via the down-regulation of E2F1 mediated by TTP in TMZ resistant glioma. temozolomide 146-149 E2F transcription factor 1 Homo sapiens 122-126 32039031-5 2019 SF7761 cells with H3F3A gene mutation showed MGMT promoter methylation, lacked MGMT expression, and sensitivity to TMZ treatment. temozolomide 115-118 H3.3 histone A Homo sapiens 18-23 32039031-0 2019 MGMT Expression Contributes to Temozolomide Resistance in H3K27M-Mutant Diffuse Midline Gliomas. temozolomide 31-43 O-6-methylguanine-DNA methyltransferase Homo sapiens 0-4 32039031-7 2019 We confirmed in vitro that MGMT expression contributes to TMZ resistance in DMG cell lines. temozolomide 58-61 O-6-methylguanine-DNA methyltransferase Homo sapiens 27-31 31713817-0 2020 LINC00174 down-regulation decreases chemoresistance to temozolomide in human glioma cells by regulating miR-138-5p/SOX9 axis. temozolomide 55-67 long intergenic non-protein coding RNA 174 Homo sapiens 0-9 31821983-2 2020 Promising, albeit limited, activity and efficacy of temozolomide have been reported in pretreated patients with metastatic colorectal cancer bearing MGMT promoter methylation. temozolomide 52-64 O-6-methylguanine-DNA methyltransferase Homo sapiens 149-153 31821983-3 2020 MGMT silencing and proficiency of the mismatch repair system were considered the major predictive biomarkers of sensitivity to temozolomide. temozolomide 127-139 O-6-methylguanine-DNA methyltransferase Homo sapiens 0-4 31713817-5 2020 Our results demonstrated that the expression level of LINC00174 was higher in glioma tissues, and LINC00174 down-regulation could remarkably prevent cell proliferation and promote cell apoptosis in both glioma cells and TMZ-resistant glioma cells. temozolomide 220-223 long intergenic non-protein coding RNA 174 Homo sapiens 54-63 31713817-5 2020 Our results demonstrated that the expression level of LINC00174 was higher in glioma tissues, and LINC00174 down-regulation could remarkably prevent cell proliferation and promote cell apoptosis in both glioma cells and TMZ-resistant glioma cells. temozolomide 220-223 long intergenic non-protein coding RNA 174 Homo sapiens 98-107 31810938-0 2019 Temozolomide-resistant Glioblastoma Depends on HDAC6 Activity Through Regulation of DNA Mismatch Repair. temozolomide 0-12 histone deacetylase 6 Homo sapiens 47-52 31964274-2 2020 Temozolomide is typically used in glioblastoma treatment; however, the enzymes alkylpurine-DNA-N-glycosylase (APNG) and methylguanine-DNA-methyltransferase (MGMT) efficiently mediate the repair of DNA damage caused by temozolomide, reducing treatment efficacy. temozolomide 0-12 N-methylpurine DNA glycosylase Homo sapiens 79-108 31964274-2 2020 Temozolomide is typically used in glioblastoma treatment; however, the enzymes alkylpurine-DNA-N-glycosylase (APNG) and methylguanine-DNA-methyltransferase (MGMT) efficiently mediate the repair of DNA damage caused by temozolomide, reducing treatment efficacy. temozolomide 0-12 N-methylpurine DNA glycosylase Homo sapiens 110-114 31964274-2 2020 Temozolomide is typically used in glioblastoma treatment; however, the enzymes alkylpurine-DNA-N-glycosylase (APNG) and methylguanine-DNA-methyltransferase (MGMT) efficiently mediate the repair of DNA damage caused by temozolomide, reducing treatment efficacy. temozolomide 0-12 O-6-methylguanine-DNA methyltransferase Homo sapiens 120-155 31964274-2 2020 Temozolomide is typically used in glioblastoma treatment; however, the enzymes alkylpurine-DNA-N-glycosylase (APNG) and methylguanine-DNA-methyltransferase (MGMT) efficiently mediate the repair of DNA damage caused by temozolomide, reducing treatment efficacy. temozolomide 0-12 O-6-methylguanine-DNA methyltransferase Homo sapiens 157-161 31964274-2 2020 Temozolomide is typically used in glioblastoma treatment; however, the enzymes alkylpurine-DNA-N-glycosylase (APNG) and methylguanine-DNA-methyltransferase (MGMT) efficiently mediate the repair of DNA damage caused by temozolomide, reducing treatment efficacy. temozolomide 218-230 N-methylpurine DNA glycosylase Homo sapiens 79-108 31964274-2 2020 Temozolomide is typically used in glioblastoma treatment; however, the enzymes alkylpurine-DNA-N-glycosylase (APNG) and methylguanine-DNA-methyltransferase (MGMT) efficiently mediate the repair of DNA damage caused by temozolomide, reducing treatment efficacy. temozolomide 218-230 N-methylpurine DNA glycosylase Homo sapiens 110-114 31964274-2 2020 Temozolomide is typically used in glioblastoma treatment; however, the enzymes alkylpurine-DNA-N-glycosylase (APNG) and methylguanine-DNA-methyltransferase (MGMT) efficiently mediate the repair of DNA damage caused by temozolomide, reducing treatment efficacy. temozolomide 218-230 O-6-methylguanine-DNA methyltransferase Homo sapiens 120-155 31964274-2 2020 Temozolomide is typically used in glioblastoma treatment; however, the enzymes alkylpurine-DNA-N-glycosylase (APNG) and methylguanine-DNA-methyltransferase (MGMT) efficiently mediate the repair of DNA damage caused by temozolomide, reducing treatment efficacy. temozolomide 218-230 O-6-methylguanine-DNA methyltransferase Homo sapiens 157-161 31906036-0 2019 Histone 2A Family Member J Drives Mesenchymal Transition and Temozolomide Resistance in Glioblastoma Multiforme. temozolomide 61-73 H2A.J histone Homo sapiens 0-26 31906036-3 2019 By using The Cancer Genome Atlas (TCGA) database, we found that the upregulation of histone 2A family member J (H2AFJ), but not other H2AFs, is extensively detected in the therapeutic-insensitive mesenchymal, IDH wildtype, MGMT unmethylated, or non-G-CIMP GBM and is associated with poor TMZ responsiveness independent of radiation. temozolomide 288-291 H2A.J histone Homo sapiens 84-110 31906036-3 2019 By using The Cancer Genome Atlas (TCGA) database, we found that the upregulation of histone 2A family member J (H2AFJ), but not other H2AFs, is extensively detected in the therapeutic-insensitive mesenchymal, IDH wildtype, MGMT unmethylated, or non-G-CIMP GBM and is associated with poor TMZ responsiveness independent of radiation. temozolomide 288-291 H2A.J histone Homo sapiens 112-117 31906036-5 2019 Whereas H2AFJ knockdown diminished TMZ resistance, H2AFJ overexpression promoted TMZ resistance in a panel of GBM cell lines. temozolomide 81-84 H2A.J histone Homo sapiens 51-56 31906036-8 2019 Moreover, we found that therapeutic targeting HADC3 by tacedinaline or NF-kappaB by ML029 is likely able to overcome the TMZ resistance in GBM cells with H2AFJ upregulation. temozolomide 121-124 nuclear factor kappa B subunit 1 Homo sapiens 71-80 31906036-8 2019 Moreover, we found that therapeutic targeting HADC3 by tacedinaline or NF-kappaB by ML029 is likely able to overcome the TMZ resistance in GBM cells with H2AFJ upregulation. temozolomide 121-124 H2A.J histone Homo sapiens 154-159 31906036-9 2019 Significantly, the GBM cohorts harboring a high-level H2AFJ transcript combined with high-level expression of TNF-alpha/NF-kappaB geneset, IL-6/STAT3 geneset or HADC3 were associated with a shorter time to tumor repopulation after initial treatment with TMZ. temozolomide 254-257 H2A.J histone Homo sapiens 54-59 31906036-10 2019 These findings not only provide H2AFJ as a biomarker to predict TMZ therapeutic effectiveness but also suggest a new strategy to combat TMZ-insensitive GBM by targeting the interaction network constructed by TNF-alpha/NF-kappaB, IL-6/STAT3, HDAC3, and H2AFJ. temozolomide 64-67 H2A.J histone Homo sapiens 32-37 31906036-10 2019 These findings not only provide H2AFJ as a biomarker to predict TMZ therapeutic effectiveness but also suggest a new strategy to combat TMZ-insensitive GBM by targeting the interaction network constructed by TNF-alpha/NF-kappaB, IL-6/STAT3, HDAC3, and H2AFJ. temozolomide 64-67 tumor necrosis factor Homo sapiens 208-217 31906036-10 2019 These findings not only provide H2AFJ as a biomarker to predict TMZ therapeutic effectiveness but also suggest a new strategy to combat TMZ-insensitive GBM by targeting the interaction network constructed by TNF-alpha/NF-kappaB, IL-6/STAT3, HDAC3, and H2AFJ. temozolomide 64-67 interleukin 6 Homo sapiens 229-233 31906036-10 2019 These findings not only provide H2AFJ as a biomarker to predict TMZ therapeutic effectiveness but also suggest a new strategy to combat TMZ-insensitive GBM by targeting the interaction network constructed by TNF-alpha/NF-kappaB, IL-6/STAT3, HDAC3, and H2AFJ. temozolomide 64-67 signal transducer and activator of transcription 3 Homo sapiens 234-239 31906036-10 2019 These findings not only provide H2AFJ as a biomarker to predict TMZ therapeutic effectiveness but also suggest a new strategy to combat TMZ-insensitive GBM by targeting the interaction network constructed by TNF-alpha/NF-kappaB, IL-6/STAT3, HDAC3, and H2AFJ. temozolomide 64-67 histone deacetylase 3 Homo sapiens 241-246 31906036-10 2019 These findings not only provide H2AFJ as a biomarker to predict TMZ therapeutic effectiveness but also suggest a new strategy to combat TMZ-insensitive GBM by targeting the interaction network constructed by TNF-alpha/NF-kappaB, IL-6/STAT3, HDAC3, and H2AFJ. temozolomide 64-67 H2A.J histone Homo sapiens 252-257 31906036-10 2019 These findings not only provide H2AFJ as a biomarker to predict TMZ therapeutic effectiveness but also suggest a new strategy to combat TMZ-insensitive GBM by targeting the interaction network constructed by TNF-alpha/NF-kappaB, IL-6/STAT3, HDAC3, and H2AFJ. temozolomide 136-139 tumor necrosis factor Homo sapiens 208-217 31906036-10 2019 These findings not only provide H2AFJ as a biomarker to predict TMZ therapeutic effectiveness but also suggest a new strategy to combat TMZ-insensitive GBM by targeting the interaction network constructed by TNF-alpha/NF-kappaB, IL-6/STAT3, HDAC3, and H2AFJ. temozolomide 136-139 interleukin 6 Homo sapiens 229-233 31906036-10 2019 These findings not only provide H2AFJ as a biomarker to predict TMZ therapeutic effectiveness but also suggest a new strategy to combat TMZ-insensitive GBM by targeting the interaction network constructed by TNF-alpha/NF-kappaB, IL-6/STAT3, HDAC3, and H2AFJ. temozolomide 136-139 signal transducer and activator of transcription 3 Homo sapiens 234-239 31906036-10 2019 These findings not only provide H2AFJ as a biomarker to predict TMZ therapeutic effectiveness but also suggest a new strategy to combat TMZ-insensitive GBM by targeting the interaction network constructed by TNF-alpha/NF-kappaB, IL-6/STAT3, HDAC3, and H2AFJ. temozolomide 136-139 histone deacetylase 3 Homo sapiens 241-246 31906036-10 2019 These findings not only provide H2AFJ as a biomarker to predict TMZ therapeutic effectiveness but also suggest a new strategy to combat TMZ-insensitive GBM by targeting the interaction network constructed by TNF-alpha/NF-kappaB, IL-6/STAT3, HDAC3, and H2AFJ. temozolomide 136-139 H2A.J histone Homo sapiens 252-257 31921673-1 2019 The Pediatric Preclinical Testing Program previously identified the PARP inhibitor talazoparib (TLZ) as a means to potentiate temozolomide (TMZ) activity for the treatment of Ewing sarcoma. temozolomide 126-138 poly(ADP-ribose) polymerase 1 Homo sapiens 68-72 31921673-1 2019 The Pediatric Preclinical Testing Program previously identified the PARP inhibitor talazoparib (TLZ) as a means to potentiate temozolomide (TMZ) activity for the treatment of Ewing sarcoma. temozolomide 140-143 poly(ADP-ribose) polymerase 1 Homo sapiens 68-72 31791385-13 2019 CONCLUSIONS: The two novel EZH2i MC4040 and MC4041 impaired primary GBM cell viability, showing even stronger effects in combination with TMZ. temozolomide 138-141 enhancer of zeste 2 polycomb repressive complex 2 subunit Homo sapiens 27-31 31889239-10 2020 To our knowledge, ours is the second study (Hoffman et al., J Neurooncol 145: 321-328, 2019) to evaluate the prognostic impact of EGFR CN gain specifically in young adults with IDH-WT GBM and in the era of modern radiation and Temozolomide, but is the first one to compares this impact with a population of adults over 45, and correlates this date with clinical onset, dimension and localization of disease between this groups. temozolomide 227-239 epidermal growth factor receptor Homo sapiens 130-134 31810002-0 2020 Inhibition of Casein Kinase II by CX-4945, But Not Yes-associated protein (YAP) by Verteporfin, Enhances the Antitumor Efficacy of Temozolomide in Glioblastoma. temozolomide 131-143 Yes1 associated transcriptional regulator Homo sapiens 75-78 31810002-3 2020 O6-methylguanine-DNA-methyltransferase (MGMT) overexpression is associated with TMZ resistance, and low MGMT is a positive response marker for TMZ therapy. temozolomide 80-83 O-6-methylguanine-DNA methyltransferase Homo sapiens 0-38 31810002-3 2020 O6-methylguanine-DNA-methyltransferase (MGMT) overexpression is associated with TMZ resistance, and low MGMT is a positive response marker for TMZ therapy. temozolomide 80-83 O-6-methylguanine-DNA methyltransferase Homo sapiens 40-44 31810002-3 2020 O6-methylguanine-DNA-methyltransferase (MGMT) overexpression is associated with TMZ resistance, and low MGMT is a positive response marker for TMZ therapy. temozolomide 143-146 O-6-methylguanine-DNA methyltransferase Homo sapiens 0-38 31810002-3 2020 O6-methylguanine-DNA-methyltransferase (MGMT) overexpression is associated with TMZ resistance, and low MGMT is a positive response marker for TMZ therapy. temozolomide 143-146 O-6-methylguanine-DNA methyltransferase Homo sapiens 104-108 31810002-5 2020 We found TMZ sensitivity is positively correlated with MGMT expression and multidrug-resistance protein ABC subfamily G member 2 (ABCG2) in these cells. temozolomide 9-12 O-6-methylguanine-DNA methyltransferase Homo sapiens 55-59 31810002-5 2020 We found TMZ sensitivity is positively correlated with MGMT expression and multidrug-resistance protein ABC subfamily G member 2 (ABCG2) in these cells. temozolomide 9-12 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 104-128 31810002-5 2020 We found TMZ sensitivity is positively correlated with MGMT expression and multidrug-resistance protein ABC subfamily G member 2 (ABCG2) in these cells. temozolomide 9-12 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 130-135 31878276-6 2019 Depletion of CD11b+Ly6G-Ly6C- MDCs in mice bearing ALTS1C1-tk tumors and receiving ganciclovir (GCV) prolonged the mean survival time for mice from 30.7 to 37.8 days, but not the controls, while the effectiveness of temozolomide was enhanced. temozolomide 216-228 integrin alpha M Mus musculus 13-18 31878276-6 2019 Depletion of CD11b+Ly6G-Ly6C- MDCs in mice bearing ALTS1C1-tk tumors and receiving ganciclovir (GCV) prolonged the mean survival time for mice from 30.7 to 37.8 days, but not the controls, while the effectiveness of temozolomide was enhanced. temozolomide 216-228 lymphocyte antigen 6 complex, locus G Mus musculus 19-23 31878276-6 2019 Depletion of CD11b+Ly6G-Ly6C- MDCs in mice bearing ALTS1C1-tk tumors and receiving ganciclovir (GCV) prolonged the mean survival time for mice from 30.7 to 37.8 days, but not the controls, while the effectiveness of temozolomide was enhanced. temozolomide 216-228 lymphocyte antigen 6 complex, locus C1 Mus musculus 24-28 31810938-6 2019 RESULTS: We showed that HDAC6-selecitve inhibitors block activation of the EGFR and p53 pathways in TMZ-resistant GBM cells. temozolomide 100-103 histone deacetylase 6 Homo sapiens 24-29 31810938-6 2019 RESULTS: We showed that HDAC6-selecitve inhibitors block activation of the EGFR and p53 pathways in TMZ-resistant GBM cells. temozolomide 100-103 epidermal growth factor receptor Homo sapiens 75-79 31810938-6 2019 RESULTS: We showed that HDAC6-selecitve inhibitors block activation of the EGFR and p53 pathways in TMZ-resistant GBM cells. temozolomide 100-103 tumor protein p53 Homo sapiens 84-87 31810938-7 2019 Importantly, the inhibition of HDAC6 correlates with increased levels of MSH2 and MSH6, key DNA mismatch repair proteins, in TMZ-resistant GBM cells. temozolomide 125-128 histone deacetylase 6 Homo sapiens 31-36 31810938-7 2019 Importantly, the inhibition of HDAC6 correlates with increased levels of MSH2 and MSH6, key DNA mismatch repair proteins, in TMZ-resistant GBM cells. temozolomide 125-128 mutS homolog 2 Homo sapiens 73-77 31810938-7 2019 Importantly, the inhibition of HDAC6 correlates with increased levels of MSH2 and MSH6, key DNA mismatch repair proteins, in TMZ-resistant GBM cells. temozolomide 125-128 mutS homolog 6 Homo sapiens 82-86 31810938-8 2019 In addition to the MSH, HDAC6 inhibitors decrease MGMT expression in TMZ-resistant GBM cells. temozolomide 69-72 msh homeobox 2 Homo sapiens 19-22 31810938-8 2019 In addition to the MSH, HDAC6 inhibitors decrease MGMT expression in TMZ-resistant GBM cells. temozolomide 69-72 histone deacetylase 6 Homo sapiens 24-29 31810938-8 2019 In addition to the MSH, HDAC6 inhibitors decrease MGMT expression in TMZ-resistant GBM cells. temozolomide 69-72 O-6-methylguanine-DNA methyltransferase Homo sapiens 50-54 31810938-9 2019 Furthermore, HDAC6 inhibitors increase TMZ sensitivity and efficiently induce apoptosis in TMZ-resistant GBM cells. temozolomide 39-42 histone deacetylase 6 Homo sapiens 13-18 31810938-9 2019 Furthermore, HDAC6 inhibitors increase TMZ sensitivity and efficiently induce apoptosis in TMZ-resistant GBM cells. temozolomide 91-94 histone deacetylase 6 Homo sapiens 13-18 31810938-10 2019 CONCLUSION: Selective inhibition of HDAC6 may be a promising strategy for the treatment of TMZ-resistant GBM. temozolomide 91-94 histone deacetylase 6 Homo sapiens 36-41 31770107-1 2019 OBJECTIVE: This study was conducted to elucidate the long non-coding RNA FOXD2-AS1 (lncRNA FOXD2-AS1) expression in glioma and its mechanism on the biological features of glioma cells and the drug resistance of temozolomide (TMZ). temozolomide 211-223 forkhead box D2 Mus musculus 73-78 31725331-0 2019 SNAP reverses temozolomide resistance in human glioblastoma multiforme cells through down-regulation of MGMT. temozolomide 14-26 O-6-methylguanine-DNA methyltransferase Homo sapiens 104-108 31725331-3 2019 Down-regulation of O-6-methylguanine-DNA methyltransferase (MGMT) expression in GBM cells is an attractive strategy for overcoming TMZ resistance and improving outcomes. temozolomide 131-134 O-6-methylguanine-DNA methyltransferase Homo sapiens 19-58 31725331-3 2019 Down-regulation of O-6-methylguanine-DNA methyltransferase (MGMT) expression in GBM cells is an attractive strategy for overcoming TMZ resistance and improving outcomes. temozolomide 131-134 O-6-methylguanine-DNA methyltransferase Homo sapiens 60-64 31725331-6 2019 SNAP acted principally through post-translational modification of p53, phosphorylated N-myc downstream regulated gene 1, and MGMT protein stability in TMZ-R GBM cells. temozolomide 151-154 O-6-methylguanine-DNA methyltransferase Homo sapiens 125-129 31725331-9 2019 SNAP reverses temozolomide resistance in human glioblastoma multiforme cells through down-regulation of MGMT. temozolomide 14-26 O-6-methylguanine-DNA methyltransferase Homo sapiens 104-108 31783653-0 2019 On the Critical Issues in Temozolomide Research in Glioblastoma: Clinically Relevant Concentrations and MGMT-independent Resistance. temozolomide 26-38 O-6-methylguanine-DNA methyltransferase Homo sapiens 104-108 31783653-8 2019 However, the clinically relevant concentrations of TMZ are sufficient to radiosensitize both MGMT-positive and -negative cell lines in vitro. temozolomide 51-54 O-6-methylguanine-DNA methyltransferase Homo sapiens 93-97 31783653-9 2019 It is speculated that a single DNA repair protein, MGMT, is highly efficient in protecting cells against TMZ toxicity. temozolomide 105-108 O-6-methylguanine-DNA methyltransferase Homo sapiens 51-55 31783653-10 2019 However, an endogenous level of MGMT protein expression is not universally correlated with TMZ responsiveness, and MGMT-independent mechanisms of TMZ resistance exist. temozolomide 146-149 O-6-methylguanine-DNA methyltransferase Homo sapiens 115-119 31639772-11 2019 Temozolomide therapy was well tolerated and most toxicities were limited to grade G1-2 according to WHO criteria. temozolomide 0-12 proline rich protein BstNI subfamily 3 Homo sapiens 82-86 31721137-0 2019 Is MGMT the best marker to predict response of temozolomide in aggressive pituitary tumors? temozolomide 47-59 O-6-methylguanine-DNA methyltransferase Homo sapiens 3-7 31807171-8 2019 The stimulation of glioma cells by IGF1 in vitro was found to decreased the viability of the cells following treatment with temozolomide (TMZ). temozolomide 124-136 insulin like growth factor 1 Homo sapiens 35-39 31807171-8 2019 The stimulation of glioma cells by IGF1 in vitro was found to decreased the viability of the cells following treatment with temozolomide (TMZ). temozolomide 138-141 insulin like growth factor 1 Homo sapiens 35-39 31807171-9 2019 In addition, the expression level of IGF1R was increased in glioma cells treated with TMZ. temozolomide 86-89 insulin like growth factor 1 receptor Homo sapiens 37-42 31571099-2 2019 Administration of TMZ results in clinical response and improvement in survival of many of these patients depending upon the expression of the DNA repair enzyme O-6 methylguanine DNA transferase (MGMT). temozolomide 18-21 O-6-methylguanine-DNA methyltransferase Homo sapiens 195-199 31571099-3 2019 Low or negative MGMT immunoreactivity predicts responsiveness to TMZ therapy. temozolomide 65-68 O-6-methylguanine-DNA methyltransferase Homo sapiens 16-20 31571099-8 2019 CONCLUSIONS: These findings could explain lack of response to TMZ treatment in patients with false negative MGMT immunohistochemistry. temozolomide 62-65 O-6-methylguanine-DNA methyltransferase Homo sapiens 108-112 31571099-9 2019 Evaluation of tumor samples for MGMT expression should carefully be carried-out using the optimum immunohistochemical protocol to obtain consistent and reliable results that help to identify patients that could respond to TMZ therapy. temozolomide 222-225 O-6-methylguanine-DNA methyltransferase Homo sapiens 32-36 31770107-1 2019 OBJECTIVE: This study was conducted to elucidate the long non-coding RNA FOXD2-AS1 (lncRNA FOXD2-AS1) expression in glioma and its mechanism on the biological features of glioma cells and the drug resistance of temozolomide (TMZ). temozolomide 211-223 arylsulfatase B Mus musculus 79-82 31770107-1 2019 OBJECTIVE: This study was conducted to elucidate the long non-coding RNA FOXD2-AS1 (lncRNA FOXD2-AS1) expression in glioma and its mechanism on the biological features of glioma cells and the drug resistance of temozolomide (TMZ). temozolomide 211-223 forkhead box D2 Mus musculus 73-82 31770107-1 2019 OBJECTIVE: This study was conducted to elucidate the long non-coding RNA FOXD2-AS1 (lncRNA FOXD2-AS1) expression in glioma and its mechanism on the biological features of glioma cells and the drug resistance of temozolomide (TMZ). temozolomide 225-228 forkhead box D2 Mus musculus 73-78 31770107-1 2019 OBJECTIVE: This study was conducted to elucidate the long non-coding RNA FOXD2-AS1 (lncRNA FOXD2-AS1) expression in glioma and its mechanism on the biological features of glioma cells and the drug resistance of temozolomide (TMZ). temozolomide 225-228 arylsulfatase B Mus musculus 79-82 31770107-1 2019 OBJECTIVE: This study was conducted to elucidate the long non-coding RNA FOXD2-AS1 (lncRNA FOXD2-AS1) expression in glioma and its mechanism on the biological features of glioma cells and the drug resistance of temozolomide (TMZ). temozolomide 225-228 forkhead box D2 Mus musculus 73-82 31771235-1 2019 Recently, we found that temozolomide (TMZ) can upregulate the expression of the multidrug-resistance protein ABCC3 in NK cells from both glioma-bearing mice and glioblastoma patients treated with dendritic cell immunotherapy combined with TMZ, allowing NK cells to escape apoptosis and favoring their role as antitumor effector cells. temozolomide 24-36 ATP-binding cassette, sub-family C (CFTR/MRP), member 3 Mus musculus 109-114 31771235-1 2019 Recently, we found that temozolomide (TMZ) can upregulate the expression of the multidrug-resistance protein ABCC3 in NK cells from both glioma-bearing mice and glioblastoma patients treated with dendritic cell immunotherapy combined with TMZ, allowing NK cells to escape apoptosis and favoring their role as antitumor effector cells. temozolomide 38-41 ATP-binding cassette, sub-family C (CFTR/MRP), member 3 Mus musculus 109-114 31771235-1 2019 Recently, we found that temozolomide (TMZ) can upregulate the expression of the multidrug-resistance protein ABCC3 in NK cells from both glioma-bearing mice and glioblastoma patients treated with dendritic cell immunotherapy combined with TMZ, allowing NK cells to escape apoptosis and favoring their role as antitumor effector cells. temozolomide 239-242 ATP-binding cassette, sub-family C (CFTR/MRP), member 3 Mus musculus 109-114 31819508-0 2019 Induction Of XLF And 53BP1 Expression Is Associated With Temozolomide Resistance In Glioblastoma Cells. temozolomide 57-69 tumor protein p53 binding protein 1 Homo sapiens 21-26 31771235-4 2019 Notably, not only basal but also TMZ-induced ABCC3 expression was related to a strong, long-term NK cell response and a better prognosis of patients. temozolomide 33-36 ATP binding cassette subfamily C member 3 Homo sapiens 45-50 31819508-7 2019 The TMZ-induced apoptosis was detected by the Caspase 3/7 activity kit. temozolomide 4-7 caspase 3 Homo sapiens 46-55 31819508-12 2019 Canonical NHEJ key factors, XLF and 53BP1, are upregulated in TMZ-resistant GBM cells. temozolomide 62-65 tumor protein p53 binding protein 1 Homo sapiens 36-41 31771235-6 2019 ABCC3 expression in patients carrying DelC compared to patients with reference haplotype was higher and modulated by TMZ. temozolomide 117-120 ATP binding cassette subfamily C member 3 Homo sapiens 0-5 31819508-13 2019 Depletion of XLF or 53BP1 in TMZ-resistant cells significantly improve the potency of TMZ against GBM cell growth. temozolomide 29-32 tumor protein p53 binding protein 1 Homo sapiens 20-25 31819508-13 2019 Depletion of XLF or 53BP1 in TMZ-resistant cells significantly improve the potency of TMZ against GBM cell growth. temozolomide 86-89 tumor protein p53 binding protein 1 Homo sapiens 20-25 31771235-7 2019 The transcription factor NRF2, involved in ABCC3 induction, was phosphorylated in CD56dim CD16+ NK cells expressing ABCC3 under TMZ treatment. temozolomide 128-131 NFE2 like bZIP transcription factor 2 Homo sapiens 25-29 31771235-7 2019 The transcription factor NRF2, involved in ABCC3 induction, was phosphorylated in CD56dim CD16+ NK cells expressing ABCC3 under TMZ treatment. temozolomide 128-131 ATP binding cassette subfamily C member 3 Homo sapiens 43-48 31771235-7 2019 The transcription factor NRF2, involved in ABCC3 induction, was phosphorylated in CD56dim CD16+ NK cells expressing ABCC3 under TMZ treatment. temozolomide 128-131 neural cell adhesion molecule 1 Homo sapiens 82-86 31771235-7 2019 The transcription factor NRF2, involved in ABCC3 induction, was phosphorylated in CD56dim CD16+ NK cells expressing ABCC3 under TMZ treatment. temozolomide 128-131 Fc gamma receptor IIIa Homo sapiens 90-94 31771235-7 2019 The transcription factor NRF2, involved in ABCC3 induction, was phosphorylated in CD56dim CD16+ NK cells expressing ABCC3 under TMZ treatment. temozolomide 128-131 ATP binding cassette subfamily C member 3 Homo sapiens 116-121 31798454-0 2019 Corrigendum: SCD1 Confers Temozolomide Resistance to Human Glioma Cells Via the Akt/GSK3beta/beta-Catenin Signaling Axis. temozolomide 26-38 stearoyl-CoA desaturase Homo sapiens 13-17 31542479-0 2019 DHFR/TYMS are positive regulators of glioma cell growth and modulate chemo-sensitivity to temozolomide. temozolomide 90-102 dihydrofolate reductase Homo sapiens 0-4 31542479-0 2019 DHFR/TYMS are positive regulators of glioma cell growth and modulate chemo-sensitivity to temozolomide. temozolomide 90-102 thymidylate synthetase Homo sapiens 5-9 31542479-5 2019 Notably, inhibition of DHFR/TYMS by pemetrexed exhibited synergistic anti-glioma activity with TMZ in both cell lines and U251 xenografts, which suggested potential combined chemotherapy for glioma. temozolomide 95-98 dihydrofolate reductase Homo sapiens 23-27 31542479-6 2019 Mechanistically, the synergistic effect of inhibition of DHFR/TYMS with TMZ was due to activated AMPK and subsequently suppressed mTOR signaling pathway. temozolomide 72-75 dihydrofolate reductase Homo sapiens 57-61 31542479-6 2019 Mechanistically, the synergistic effect of inhibition of DHFR/TYMS with TMZ was due to activated AMPK and subsequently suppressed mTOR signaling pathway. temozolomide 72-75 thymidylate synthetase Homo sapiens 62-66 31542479-7 2019 Taken together, these findings identify an uncharacterized role of DHFR/TYMS in glioma growth and TMZ sensitivity mediated by AMPK-mTOR signal pathway, and provide a prospective approach for improving the anti-tumor activity of TMZ in glioma. temozolomide 98-101 dihydrofolate reductase Homo sapiens 67-71 31542479-7 2019 Taken together, these findings identify an uncharacterized role of DHFR/TYMS in glioma growth and TMZ sensitivity mediated by AMPK-mTOR signal pathway, and provide a prospective approach for improving the anti-tumor activity of TMZ in glioma. temozolomide 98-101 thymidylate synthetase Homo sapiens 72-76 31542479-7 2019 Taken together, these findings identify an uncharacterized role of DHFR/TYMS in glioma growth and TMZ sensitivity mediated by AMPK-mTOR signal pathway, and provide a prospective approach for improving the anti-tumor activity of TMZ in glioma. temozolomide 228-231 dihydrofolate reductase Homo sapiens 67-71 31542479-7 2019 Taken together, these findings identify an uncharacterized role of DHFR/TYMS in glioma growth and TMZ sensitivity mediated by AMPK-mTOR signal pathway, and provide a prospective approach for improving the anti-tumor activity of TMZ in glioma. temozolomide 228-231 thymidylate synthetase Homo sapiens 72-76 31798454-0 2019 Corrigendum: SCD1 Confers Temozolomide Resistance to Human Glioma Cells Via the Akt/GSK3beta/beta-Catenin Signaling Axis. temozolomide 26-38 AKT serine/threonine kinase 1 Homo sapiens 80-83 31798454-0 2019 Corrigendum: SCD1 Confers Temozolomide Resistance to Human Glioma Cells Via the Akt/GSK3beta/beta-Catenin Signaling Axis. temozolomide 26-38 glycogen synthase kinase 3 beta Homo sapiens 84-92 31798454-0 2019 Corrigendum: SCD1 Confers Temozolomide Resistance to Human Glioma Cells Via the Akt/GSK3beta/beta-Catenin Signaling Axis. temozolomide 26-38 catenin beta 1 Homo sapiens 93-105 31814867-9 2019 Additionally, a combination assay of PP7 with temozolomide (TMZ), the most used chemotherapy for glioma patients, was performed resulting in synergism, while PP7 reduced TMZ resistance through inhibition of MGMT expression. temozolomide 60-63 protein phosphatase with EF-hand domain 1 Homo sapiens 37-40 31814867-9 2019 Additionally, a combination assay of PP7 with temozolomide (TMZ), the most used chemotherapy for glioma patients, was performed resulting in synergism, while PP7 reduced TMZ resistance through inhibition of MGMT expression. temozolomide 170-173 protein phosphatase with EF-hand domain 1 Homo sapiens 158-161 31814867-9 2019 Additionally, a combination assay of PP7 with temozolomide (TMZ), the most used chemotherapy for glioma patients, was performed resulting in synergism, while PP7 reduced TMZ resistance through inhibition of MGMT expression. temozolomide 170-173 O-6-methylguanine-DNA methyltransferase Homo sapiens 207-211 31717924-0 2019 ANGPTL4 Induces TMZ Resistance of Glioblastoma by Promoting Cancer Stemness Enrichment via the EGFR/AKT/4E-BP1 Cascade. temozolomide 16-19 angiopoietin like 4 Homo sapiens 0-7 31717973-5 2019 In experimental settings, high doses of the agent are often used, which are likely to activate responses triggered by base N-alkylations instead of O6-methylguanine (O6MeG), which is the primary cytotoxic lesion induced by low doses of temozolomide and other methylating drugs in O6-methylguanine-DNA methyltransferase (MGMT) repair incompetent cells. temozolomide 236-248 O-6-methylguanine-DNA methyltransferase Homo sapiens 280-318 31717973-5 2019 In experimental settings, high doses of the agent are often used, which are likely to activate responses triggered by base N-alkylations instead of O6-methylguanine (O6MeG), which is the primary cytotoxic lesion induced by low doses of temozolomide and other methylating drugs in O6-methylguanine-DNA methyltransferase (MGMT) repair incompetent cells. temozolomide 236-248 O-6-methylguanine-DNA methyltransferase Homo sapiens 320-324 31717973-7 2019 MGMT, repairing the lesion through methyl group transfer, is a key node in protecting cells against all these effects and has a significant impact on patient"s survival following temozolomide therapy, supporting the notion that findings obtained on a molecular and cellular level can be translated to the therapeutic setting in vivo. temozolomide 179-191 O-6-methylguanine-DNA methyltransferase Homo sapiens 0-4 31717924-0 2019 ANGPTL4 Induces TMZ Resistance of Glioblastoma by Promoting Cancer Stemness Enrichment via the EGFR/AKT/4E-BP1 Cascade. temozolomide 16-19 epidermal growth factor receptor Homo sapiens 95-99 31717924-0 2019 ANGPTL4 Induces TMZ Resistance of Glioblastoma by Promoting Cancer Stemness Enrichment via the EGFR/AKT/4E-BP1 Cascade. temozolomide 16-19 AKT serine/threonine kinase 1 Homo sapiens 100-103 31717924-0 2019 ANGPTL4 Induces TMZ Resistance of Glioblastoma by Promoting Cancer Stemness Enrichment via the EGFR/AKT/4E-BP1 Cascade. temozolomide 16-19 eukaryotic translation initiation factor 4E binding protein 1 Homo sapiens 104-110 31717924-6 2019 The overexpression of ANGPTL4 induced GSC enrichment that was characterized by polycomb complex protein BMI-1 and SRY (sex determining region Y)-box 2 (SOX2) expression, resulting in TMZ resistance in GBM. temozolomide 183-186 angiopoietin like 4 Homo sapiens 22-29 31717924-6 2019 The overexpression of ANGPTL4 induced GSC enrichment that was characterized by polycomb complex protein BMI-1 and SRY (sex determining region Y)-box 2 (SOX2) expression, resulting in TMZ resistance in GBM. temozolomide 183-186 SRY-box transcription factor 2 Homo sapiens 152-156 31680769-0 2019 FoxD2-AS1 is a prognostic factor in glioma and promotes temozolomide resistance in a O6-methylguanine-DNA methyltransferase-dependent manner. temozolomide 56-68 FOXD2 adjacent opposite strand RNA 1 Homo sapiens 0-9 31708690-7 2019 Then, we found that overexpression of p62 promoted glioma progression by promoting proliferation, migration, glycolysis, temozolomide (TMZ) resistance and nuclear factor kappaB (NF-kappaB) signalling pathway, and repressing autophagic flux and reactive oxygen species (ROS) in vitro. temozolomide 121-133 nucleoporin 62 Homo sapiens 38-41 31708690-7 2019 Then, we found that overexpression of p62 promoted glioma progression by promoting proliferation, migration, glycolysis, temozolomide (TMZ) resistance and nuclear factor kappaB (NF-kappaB) signalling pathway, and repressing autophagic flux and reactive oxygen species (ROS) in vitro. temozolomide 135-138 nucleoporin 62 Homo sapiens 38-41 31586022-1 2019 OBJECTIVE: To determine the efficacy of the thrombopoietin receptor agonist romiplostim for the prevention of temozolomide-induced thrombocytopenia in newly diagnosed glioblastoma. temozolomide 110-122 MPL proto-oncogene, thrombopoietin receptor Homo sapiens 44-67 31586022-10 2019 CONCLUSION: The thrombopoietin receptor agonist romiplostim improves exposure to chemotherapy in patients with glioblastoma experiencing temozolomide-induced thrombocytopenia. temozolomide 137-149 MPL proto-oncogene, thrombopoietin receptor Homo sapiens 16-39 31815044-0 2019 BRCA1 identified as a modulator of temozolomide resistance in P53 wild-type GBM using a high-throughput shRNA-based synthetic lethality screening. temozolomide 35-47 BRCA1 DNA repair associated Homo sapiens 0-5 31815044-0 2019 BRCA1 identified as a modulator of temozolomide resistance in P53 wild-type GBM using a high-throughput shRNA-based synthetic lethality screening. temozolomide 35-47 tumor protein p53 Homo sapiens 62-65 31815044-5 2019 Using an unbiased bioinformatical analysis, we identified BRCA1 as a potential promising candidate gene that induced synthetic lethality with TMZ in glioma sphere-forming cells (GSCs). temozolomide 142-145 BRCA1 DNA repair associated Homo sapiens 58-63 31815044-6 2019 BRCA1 knockdown resulted in antitumor activity with TMZ in P53 wild-type GSCs but not in P53 mutant GSCs. temozolomide 52-55 BRCA1 DNA repair associated Homo sapiens 0-5 31815044-6 2019 BRCA1 knockdown resulted in antitumor activity with TMZ in P53 wild-type GSCs but not in P53 mutant GSCs. temozolomide 52-55 tumor protein p53 Homo sapiens 59-62 31815044-7 2019 TMZ treatment induced a DNA damage repair response; the activation of BRCA1 DNA repair pathway targets and knockdown of BRCA1, together with TMZ, led to increased DNA damage and cell death in P53 wild-type GSCs. temozolomide 0-3 BRCA1 DNA repair associated Homo sapiens 70-75 31815044-7 2019 TMZ treatment induced a DNA damage repair response; the activation of BRCA1 DNA repair pathway targets and knockdown of BRCA1, together with TMZ, led to increased DNA damage and cell death in P53 wild-type GSCs. temozolomide 0-3 BRCA1 DNA repair associated Homo sapiens 120-125 31815044-7 2019 TMZ treatment induced a DNA damage repair response; the activation of BRCA1 DNA repair pathway targets and knockdown of BRCA1, together with TMZ, led to increased DNA damage and cell death in P53 wild-type GSCs. temozolomide 0-3 tumor protein p53 Homo sapiens 192-195 31815044-7 2019 TMZ treatment induced a DNA damage repair response; the activation of BRCA1 DNA repair pathway targets and knockdown of BRCA1, together with TMZ, led to increased DNA damage and cell death in P53 wild-type GSCs. temozolomide 141-144 tumor protein p53 Homo sapiens 192-195 31815044-8 2019 Our study identified BRCA1 as a potential target that sensitizes TMZ-induced cell death in P53 wild-type GBM, suggesting that the combined inhibition of BRCA1 and TMZ treatment will be a successful targeted therapy for GBM patients. temozolomide 65-68 BRCA1 DNA repair associated Homo sapiens 21-26 31815044-8 2019 Our study identified BRCA1 as a potential target that sensitizes TMZ-induced cell death in P53 wild-type GBM, suggesting that the combined inhibition of BRCA1 and TMZ treatment will be a successful targeted therapy for GBM patients. temozolomide 65-68 tumor protein p53 Homo sapiens 91-94 31815044-8 2019 Our study identified BRCA1 as a potential target that sensitizes TMZ-induced cell death in P53 wild-type GBM, suggesting that the combined inhibition of BRCA1 and TMZ treatment will be a successful targeted therapy for GBM patients. temozolomide 65-68 BRCA1 DNA repair associated Homo sapiens 153-158 31815044-8 2019 Our study identified BRCA1 as a potential target that sensitizes TMZ-induced cell death in P53 wild-type GBM, suggesting that the combined inhibition of BRCA1 and TMZ treatment will be a successful targeted therapy for GBM patients. temozolomide 163-166 BRCA1 DNA repair associated Homo sapiens 21-26 31815044-8 2019 Our study identified BRCA1 as a potential target that sensitizes TMZ-induced cell death in P53 wild-type GBM, suggesting that the combined inhibition of BRCA1 and TMZ treatment will be a successful targeted therapy for GBM patients. temozolomide 163-166 tumor protein p53 Homo sapiens 91-94 31614149-0 2019 Nuclear factor I A promotes temozolomide resistance in glioblastoma via activation of nuclear factor kappaB pathway. temozolomide 28-40 nuclear factor I A Homo sapiens 0-18 31614149-2 2019 MAIN METHODS: A genome-wide hierarchical bi-clustering based on previously published microarray databases identified Nuclear Factor I A (NFIA) as one of the most significantly upregulated genes correlated to TMZ resistance in GBM. temozolomide 208-211 nuclear factor I A Homo sapiens 117-135 31614149-2 2019 MAIN METHODS: A genome-wide hierarchical bi-clustering based on previously published microarray databases identified Nuclear Factor I A (NFIA) as one of the most significantly upregulated genes correlated to TMZ resistance in GBM. temozolomide 208-211 nuclear factor I A Homo sapiens 137-141 31614149-3 2019 Then, the potential biological functions of NFIA in oncogenesis and chemoresistance were clarified by qRT-PCR, Western blotting and in vivo xenograft models with artificially induced TMZ-resistant U87 cells. temozolomide 183-186 nuclear factor I A Homo sapiens 44-48 31614149-6 2019 KEY FINDINGS: NFIA was correlated with TMZ resistance in GBM. temozolomide 39-42 nuclear factor I A Homo sapiens 14-18 31680769-0 2019 FoxD2-AS1 is a prognostic factor in glioma and promotes temozolomide resistance in a O6-methylguanine-DNA methyltransferase-dependent manner. temozolomide 56-68 O-6-methylguanine-DNA methyltransferase Homo sapiens 85-123 31614149-8 2019 Moreover, NFIA contributed to the acquired TMZ resistance of GBM cells, while suppression of NFIA via lentivirus reduced cell proliferation, tumorigenesis and resistance to TMZ of GBM. temozolomide 173-176 nuclear factor I A Homo sapiens 93-97 31680769-3 2019 Overexpression of long-noncoding RNA (LncRNA) FoxD2 adjacent opposite strand RNA 1 (FoxD2-AS1) was identified to promote glioma development, but the role in TMZ resistance remains unclear. temozolomide 157-160 forkhead box D2 Homo sapiens 46-51 31614149-10 2019 Last, NFIA induced phosphorylation of NF-kB p65 at serine 536, thus inducing TMZ resistance in GBM cells. temozolomide 77-80 nuclear factor I A Homo sapiens 6-10 31680769-3 2019 Overexpression of long-noncoding RNA (LncRNA) FoxD2 adjacent opposite strand RNA 1 (FoxD2-AS1) was identified to promote glioma development, but the role in TMZ resistance remains unclear. temozolomide 157-160 FOXD2 adjacent opposite strand RNA 1 Homo sapiens 84-93 31614149-10 2019 Last, NFIA induced phosphorylation of NF-kB p65 at serine 536, thus inducing TMZ resistance in GBM cells. temozolomide 77-80 RELA proto-oncogene, NF-kB subunit Homo sapiens 44-47 31614149-11 2019 Altogether, our study suggests that NFIA-dependent transcriptional regulation of NF-kB contributes to acquired TMZ resistance in GBM. temozolomide 111-114 nuclear factor I A Homo sapiens 36-40 31680769-6 2019 Knockdown of FoxD2-AS1 decreased the proliferation, metastatic ability of glioma cells and promote the sensitivity to TMZ in glioma cells. temozolomide 118-121 FOXD2 adjacent opposite strand RNA 1 Homo sapiens 13-22 31614149-12 2019 SIGNIFICANCE: Abnormally activated NFIA-NF-kB signaling was strongly correlated with acquired TMZ resistance and poor prognosis in GBM, and it could be a new therapeutic target for TMZ-resistant GBM. temozolomide 94-97 nuclear factor I A Homo sapiens 35-39 31614149-12 2019 SIGNIFICANCE: Abnormally activated NFIA-NF-kB signaling was strongly correlated with acquired TMZ resistance and poor prognosis in GBM, and it could be a new therapeutic target for TMZ-resistant GBM. temozolomide 181-184 nuclear factor I A Homo sapiens 35-39 31680769-8 2019 Our data suggested that FoxD2-AS1 is a clinical relevance LncRNA and mediates TMZ resistance by regulating the methylation status of the MGMT promoter region. temozolomide 78-81 forkhead box D2 Homo sapiens 24-29 31680769-8 2019 Our data suggested that FoxD2-AS1 is a clinical relevance LncRNA and mediates TMZ resistance by regulating the methylation status of the MGMT promoter region. temozolomide 78-81 prostaglandin D2 receptor Homo sapiens 30-33 31680769-8 2019 Our data suggested that FoxD2-AS1 is a clinical relevance LncRNA and mediates TMZ resistance by regulating the methylation status of the MGMT promoter region. temozolomide 78-81 O-6-methylguanine-DNA methyltransferase Homo sapiens 137-141 31563286-0 2019 Targeting Aurora kinase B attenuates chemoresistance in glioblastoma via a synergistic manner with temozolomide. temozolomide 99-111 aurora kinase B Homo sapiens 10-25 31563286-3 2019 In this study, we investigate the underlying mechanism by which glioblastoma (GBM) cells acquire resistance to Temozolomide (TMZ) through Aurora kinase B (AURKB) thus to identify novel therapeutic targets and prognostic biomarkers for GBM. temozolomide 111-123 aurora kinase B Homo sapiens 138-153 31563286-3 2019 In this study, we investigate the underlying mechanism by which glioblastoma (GBM) cells acquire resistance to Temozolomide (TMZ) through Aurora kinase B (AURKB) thus to identify novel therapeutic targets and prognostic biomarkers for GBM. temozolomide 111-123 aurora kinase B Homo sapiens 155-160 31563286-3 2019 In this study, we investigate the underlying mechanism by which glioblastoma (GBM) cells acquire resistance to Temozolomide (TMZ) through Aurora kinase B (AURKB) thus to identify novel therapeutic targets and prognostic biomarkers for GBM. temozolomide 125-128 aurora kinase B Homo sapiens 138-153 31563286-3 2019 In this study, we investigate the underlying mechanism by which glioblastoma (GBM) cells acquire resistance to Temozolomide (TMZ) through Aurora kinase B (AURKB) thus to identify novel therapeutic targets and prognostic biomarkers for GBM. temozolomide 125-128 aurora kinase B Homo sapiens 155-160 31563286-8 2019 RESULTS: AURKB was among the most significantly up-regulated kinase-coding genes in TMZ resistant GBM cells according to database GSE68029, moreover, an increased expression of AURKB was closely associated with poor prognosis in glioma and GBM patients. temozolomide 84-87 aurora kinase B Homo sapiens 9-14 31563286-8 2019 RESULTS: AURKB was among the most significantly up-regulated kinase-coding genes in TMZ resistant GBM cells according to database GSE68029, moreover, an increased expression of AURKB was closely associated with poor prognosis in glioma and GBM patients. temozolomide 84-87 aurora kinase B Homo sapiens 177-182 31563286-9 2019 AURKB knock-down resensitized U87 resistant cells to TMZ both in vitro and in vivo. temozolomide 53-56 aurora kinase B Homo sapiens 0-5 31563286-10 2019 Additionally, the combination of TMZ and Hesperadin, a specific AURKB inhibitor, significantly suppressed the proliferation of TMZ resistant GBM cells thus dramatically prolonged the survival of xenograft mice viaa synergistic effect with TMZ. temozolomide 33-36 aurora kinase B Mus musculus 64-69 31563286-10 2019 Additionally, the combination of TMZ and Hesperadin, a specific AURKB inhibitor, significantly suppressed the proliferation of TMZ resistant GBM cells thus dramatically prolonged the survival of xenograft mice viaa synergistic effect with TMZ. temozolomide 127-130 aurora kinase B Mus musculus 64-69 31563286-10 2019 Additionally, the combination of TMZ and Hesperadin, a specific AURKB inhibitor, significantly suppressed the proliferation of TMZ resistant GBM cells thus dramatically prolonged the survival of xenograft mice viaa synergistic effect with TMZ. temozolomide 127-130 aurora kinase B Mus musculus 64-69 31563286-11 2019 CONCLUSION: Elevated AURKB expression was strongly correlated to TMZ resistant acquisition and poor prognosis, furthermore, targeting AURKB would be a potential therapeutic target for GBM patients. temozolomide 65-68 aurora kinase B Homo sapiens 21-26 31563286-11 2019 CONCLUSION: Elevated AURKB expression was strongly correlated to TMZ resistant acquisition and poor prognosis, furthermore, targeting AURKB would be a potential therapeutic target for GBM patients. temozolomide 65-68 aurora kinase B Homo sapiens 134-139 31629402-0 2019 Enrichment of superoxide dismutase 2 in glioblastoma confers to acquisition of temozolomide resistance that is associated with tumor-initiating cell subsets. temozolomide 79-91 superoxide dismutase 2 Homo sapiens 14-36 31601097-7 2019 Besides, tFNA-TMZ was able to attenuate drug resistance in TMZ-resistant cells (T98G and LN-18) via downregulating the expression of O6-methylguanine-DNA-methyltransferase (MGMT). temozolomide 14-17 O-6-methylguanine-DNA methyltransferase Mus musculus 133-171 31601097-7 2019 Besides, tFNA-TMZ was able to attenuate drug resistance in TMZ-resistant cells (T98G and LN-18) via downregulating the expression of O6-methylguanine-DNA-methyltransferase (MGMT). temozolomide 14-17 O-6-methylguanine-DNA methyltransferase Mus musculus 173-177 31601097-7 2019 Besides, tFNA-TMZ was able to attenuate drug resistance in TMZ-resistant cells (T98G and LN-18) via downregulating the expression of O6-methylguanine-DNA-methyltransferase (MGMT). temozolomide 59-62 O-6-methylguanine-DNA methyltransferase Mus musculus 133-171 31601097-7 2019 Besides, tFNA-TMZ was able to attenuate drug resistance in TMZ-resistant cells (T98G and LN-18) via downregulating the expression of O6-methylguanine-DNA-methyltransferase (MGMT). temozolomide 59-62 O-6-methylguanine-DNA methyltransferase Mus musculus 173-177 31629402-7 2019 SOD2 in the resistant cells functionally determined the cell fate by limiting TMZ-stimulated superoxide reaction and cleavage of caspase-3. temozolomide 78-81 superoxide dismutase 2 Homo sapiens 0-4 31629402-12 2019 Finally, co-treatment with TMZ and the SOD inhibitor sodium diethyldithiocarbamate trihydrate in xenograft mouse models with the TMZ-resistant primary tumor resulted in lower tumor proliferation, longer survival, and less CD133, Bmi-1, and SOD2 expression. temozolomide 27-30 prominin 1 Mus musculus 222-227 31629402-12 2019 Finally, co-treatment with TMZ and the SOD inhibitor sodium diethyldithiocarbamate trihydrate in xenograft mouse models with the TMZ-resistant primary tumor resulted in lower tumor proliferation, longer survival, and less CD133, Bmi-1, and SOD2 expression. temozolomide 27-30 Bmi1 polycomb ring finger oncogene Mus musculus 229-234 31639020-0 2019 The Chk1 inhibitor SAR-020106 sensitizes human glioblastoma cells to irradiation, to temozolomide, and to decitabine treatment. temozolomide 85-97 checkpoint kinase 1 Homo sapiens 4-8 31639020-0 2019 The Chk1 inhibitor SAR-020106 sensitizes human glioblastoma cells to irradiation, to temozolomide, and to decitabine treatment. temozolomide 85-97 sarcosine dehydrogenase Homo sapiens 19-29 31629402-12 2019 Finally, co-treatment with TMZ and the SOD inhibitor sodium diethyldithiocarbamate trihydrate in xenograft mouse models with the TMZ-resistant primary tumor resulted in lower tumor proliferation, longer survival, and less CD133, Bmi-1, and SOD2 expression. temozolomide 27-30 superoxide dismutase 2, mitochondrial Mus musculus 240-244 31629402-12 2019 Finally, co-treatment with TMZ and the SOD inhibitor sodium diethyldithiocarbamate trihydrate in xenograft mouse models with the TMZ-resistant primary tumor resulted in lower tumor proliferation, longer survival, and less CD133, Bmi-1, and SOD2 expression. temozolomide 129-132 prominin 1 Mus musculus 222-227 31629402-12 2019 Finally, co-treatment with TMZ and the SOD inhibitor sodium diethyldithiocarbamate trihydrate in xenograft mouse models with the TMZ-resistant primary tumor resulted in lower tumor proliferation, longer survival, and less CD133, Bmi-1, and SOD2 expression. temozolomide 129-132 Bmi1 polycomb ring finger oncogene Mus musculus 229-234 31629402-12 2019 Finally, co-treatment with TMZ and the SOD inhibitor sodium diethyldithiocarbamate trihydrate in xenograft mouse models with the TMZ-resistant primary tumor resulted in lower tumor proliferation, longer survival, and less CD133, Bmi-1, and SOD2 expression. temozolomide 129-132 superoxide dismutase 2, mitochondrial Mus musculus 240-244 31629402-13 2019 CONCLUSION: SOD2 plays crucial roles in the tumor-initiating features that are related to TMZ resistance. temozolomide 90-93 superoxide dismutase 2 Homo sapiens 12-16 31681604-7 2019 Moreover, miR-199a renders tumor cells more sensitive to temozolomide (TMZ) via targeting K-RAS. temozolomide 57-69 KRAS proto-oncogene, GTPase Homo sapiens 90-95 31681604-7 2019 Moreover, miR-199a renders tumor cells more sensitive to temozolomide (TMZ) via targeting K-RAS. temozolomide 71-74 KRAS proto-oncogene, GTPase Homo sapiens 90-95 31561595-6 2019 Bioinformatics data have indicated that dopamine receptor (DRD) 2, DRD4, CD133 and Nestin were elevated in GBM clinical samples and correlated to Temozolomide (TMZ) resistance and increased aldehyde dehydrogenase (ALDH) activity (3.5-8.9%) as well as enhanced (2.1-2.4-fold) neurosphere formation efficiency in U87MG and D54MG GBM cell lines. temozolomide 146-158 dopamine receptor D4 Homo sapiens 67-71 31632968-0 2019 Novel Function of lncRNA ADAMTS9-AS2 in Promoting Temozolomide Resistance in Glioblastoma via Upregulating the FUS/MDM2 Ubiquitination Axis. temozolomide 50-62 ADAM metallopeptidase with thrombospondin type 1 motif 9 Homo sapiens 25-32 31632968-0 2019 Novel Function of lncRNA ADAMTS9-AS2 in Promoting Temozolomide Resistance in Glioblastoma via Upregulating the FUS/MDM2 Ubiquitination Axis. temozolomide 50-62 FUS RNA binding protein Homo sapiens 111-114 31632968-0 2019 Novel Function of lncRNA ADAMTS9-AS2 in Promoting Temozolomide Resistance in Glioblastoma via Upregulating the FUS/MDM2 Ubiquitination Axis. temozolomide 50-62 MDM2 proto-oncogene Homo sapiens 115-119 31632968-4 2019 LncRNAs from four public datasets were reanalyzed, and the candidate lncRNA ADAMTS9-AS2 was evaluated in TMZ-treated GBM patients and in vitro cell lines. temozolomide 105-108 ADAM metallopeptidase with thrombospondin type 1 motif 9 Homo sapiens 76-83 31632968-5 2019 Results: Reanalysis of lncRNA expression profiles identified ADAMTS9-AS2 as significantly overexpressed in TMZ-resistant GBM cells and as positively associated with the IC50 of TMZ in GBM cells. temozolomide 107-110 ADAM metallopeptidase with thrombospondin type 1 motif 9 Homo sapiens 61-68 31632968-5 2019 Results: Reanalysis of lncRNA expression profiles identified ADAMTS9-AS2 as significantly overexpressed in TMZ-resistant GBM cells and as positively associated with the IC50 of TMZ in GBM cells. temozolomide 177-180 ADAM metallopeptidase with thrombospondin type 1 motif 9 Homo sapiens 61-68 31632968-6 2019 Overexpression of ADAMTS9-AS2 was also significantly associated with poor TMZ response and shorter progression-free survival (PFS) in TMZ-treated GBM patients. temozolomide 74-77 ADAM metallopeptidase with thrombospondin type 1 motif 9 Homo sapiens 18-25 31632968-6 2019 Overexpression of ADAMTS9-AS2 was also significantly associated with poor TMZ response and shorter progression-free survival (PFS) in TMZ-treated GBM patients. temozolomide 134-137 ADAM metallopeptidase with thrombospondin type 1 motif 9 Homo sapiens 18-25 31632968-7 2019 Knockdown of ADAMTS9-AS2 inhibited proliferation and attenuated the IC50 of TMZ, as well as mitigating invasion and migration in TMZ-resistant GBM cells. temozolomide 76-79 ADAM metallopeptidase with thrombospondin type 1 motif 9 Homo sapiens 13-20 31632968-7 2019 Knockdown of ADAMTS9-AS2 inhibited proliferation and attenuated the IC50 of TMZ, as well as mitigating invasion and migration in TMZ-resistant GBM cells. temozolomide 129-132 ADAM metallopeptidase with thrombospondin type 1 motif 9 Homo sapiens 13-20 31632968-9 2019 Expression trends of FUS were directly correlated with those of ADAMTS9-AS2, as shown by increasing concentrations and prolonged treatment with TMZ. temozolomide 144-147 FUS RNA binding protein Homo sapiens 21-24 31632968-9 2019 Expression trends of FUS were directly correlated with those of ADAMTS9-AS2, as shown by increasing concentrations and prolonged treatment with TMZ. temozolomide 144-147 ADAM metallopeptidase with thrombospondin type 1 motif 9 Homo sapiens 64-71 31632968-14 2019 Additionally, knockdown of the ADAMTS9-AS2/FUS signaling axis significantly alleviated progression and metastasis in TMZ-resistant cells. temozolomide 117-120 ADAM metallopeptidase with thrombospondin type 1 motif 9 Homo sapiens 31-38 31632968-14 2019 Additionally, knockdown of the ADAMTS9-AS2/FUS signaling axis significantly alleviated progression and metastasis in TMZ-resistant cells. temozolomide 117-120 FUS RNA binding protein Homo sapiens 43-46 31632968-15 2019 Conclusion: ADAMTS9-AS2 possessed a novel function that promotes TMZ resistance via upregulating the FUS/MDM2 axis in GBM cells. temozolomide 65-68 ADAM metallopeptidase with thrombospondin type 1 motif 9 Homo sapiens 12-19 31632968-15 2019 Conclusion: ADAMTS9-AS2 possessed a novel function that promotes TMZ resistance via upregulating the FUS/MDM2 axis in GBM cells. temozolomide 65-68 FUS RNA binding protein Homo sapiens 101-104 31632968-15 2019 Conclusion: ADAMTS9-AS2 possessed a novel function that promotes TMZ resistance via upregulating the FUS/MDM2 axis in GBM cells. temozolomide 65-68 MDM2 proto-oncogene Homo sapiens 105-109 31632968-16 2019 The RRM or Znf_RanBP2 domains of FUS facilitate the combination of ADAMTS9-AS2 and FUS, competitively inhibiting MDM2-dependent FUS K48 ubiquitination and resulting in enhanced FUS stability and TMZ resistance. temozolomide 195-198 FUS RNA binding protein Homo sapiens 33-36 31632968-16 2019 The RRM or Znf_RanBP2 domains of FUS facilitate the combination of ADAMTS9-AS2 and FUS, competitively inhibiting MDM2-dependent FUS K48 ubiquitination and resulting in enhanced FUS stability and TMZ resistance. temozolomide 195-198 ADAM metallopeptidase with thrombospondin type 1 motif 9 Homo sapiens 67-74 31632968-16 2019 The RRM or Znf_RanBP2 domains of FUS facilitate the combination of ADAMTS9-AS2 and FUS, competitively inhibiting MDM2-dependent FUS K48 ubiquitination and resulting in enhanced FUS stability and TMZ resistance. temozolomide 195-198 FUS RNA binding protein Homo sapiens 83-86 31632968-16 2019 The RRM or Znf_RanBP2 domains of FUS facilitate the combination of ADAMTS9-AS2 and FUS, competitively inhibiting MDM2-dependent FUS K48 ubiquitination and resulting in enhanced FUS stability and TMZ resistance. temozolomide 195-198 MDM2 proto-oncogene Homo sapiens 113-117 31632968-16 2019 The RRM or Znf_RanBP2 domains of FUS facilitate the combination of ADAMTS9-AS2 and FUS, competitively inhibiting MDM2-dependent FUS K48 ubiquitination and resulting in enhanced FUS stability and TMZ resistance. temozolomide 195-198 FUS RNA binding protein Homo sapiens 83-86 31632968-16 2019 The RRM or Znf_RanBP2 domains of FUS facilitate the combination of ADAMTS9-AS2 and FUS, competitively inhibiting MDM2-dependent FUS K48 ubiquitination and resulting in enhanced FUS stability and TMZ resistance. temozolomide 195-198 FUS RNA binding protein Homo sapiens 83-86 31632968-17 2019 Our results suggest that the ADAMTS9-AS2/FUS/MDM2 axis may represent a suitable prognostic biomarker and a potential target in TMZ-resistant GBM therapy. temozolomide 127-130 ADAM metallopeptidase with thrombospondin type 1 motif 9 Homo sapiens 29-36 31632968-17 2019 Our results suggest that the ADAMTS9-AS2/FUS/MDM2 axis may represent a suitable prognostic biomarker and a potential target in TMZ-resistant GBM therapy. temozolomide 127-130 FUS RNA binding protein Homo sapiens 41-44 31632968-17 2019 Our results suggest that the ADAMTS9-AS2/FUS/MDM2 axis may represent a suitable prognostic biomarker and a potential target in TMZ-resistant GBM therapy. temozolomide 127-130 MDM2 proto-oncogene Homo sapiens 45-49 31611911-0 2019 A Novel DNA Methylation-Based Signature Can Predict the Responses of MGMT Promoter Unmethylated Glioblastomas to Temozolomide. temozolomide 113-125 O-6-methylguanine-DNA methyltransferase Homo sapiens 69-73 31611911-3 2019 DNA methylation profiling holds great promise for further stratifying the responses of MGMT promoter unmethylated GBMs to TMZ. temozolomide 122-125 O-6-methylguanine-DNA methyltransferase Homo sapiens 87-91 31611911-4 2019 In this study, we studied 147 TMZ-treated MGMT promoter unmethylated GBM, whose methylation information was obtained from the HumanMethylation27 (HM-27K) BeadChips (n = 107) and the HumanMethylation450 (HM-450K) BeadChips (n = 40) for training and validation, respectively. temozolomide 30-33 O-6-methylguanine-DNA methyltransferase Homo sapiens 42-46 31611911-5 2019 In the training set, we performed univariate Cox regression and identified that 3,565 CpGs were significantly associated with the OS of the TMZ-treated MGMT promoter unmethylated GBMs. temozolomide 140-143 O-6-methylguanine-DNA methyltransferase Homo sapiens 152-156 31611911-8 2019 In both training and validation datasets, the signature identified the TMZ-sensitive GBMs in the MGMT promoter unmethylated GBMs, and only the patients in the low-risk group appear to benefit from the TMZ treatment. temozolomide 71-74 O-6-methylguanine-DNA methyltransferase Homo sapiens 97-101 31611911-8 2019 In both training and validation datasets, the signature identified the TMZ-sensitive GBMs in the MGMT promoter unmethylated GBMs, and only the patients in the low-risk group appear to benefit from the TMZ treatment. temozolomide 201-204 O-6-methylguanine-DNA methyltransferase Homo sapiens 97-101 31611911-9 2019 Furthermore, these identified TMZ-sensitive MGMT promoter unmethylated GBMs have a similar OS when compared with the MGMT promoter methylated GBMs after TMZ treatment in both two datasets. temozolomide 30-33 O-6-methylguanine-DNA methyltransferase Homo sapiens 44-48 31611911-10 2019 Multivariate Cox regression demonstrated the independent prognostic value of the signature in TMZ-treated MGMT promoter unmethylated GBMs. temozolomide 94-97 O-6-methylguanine-DNA methyltransferase Homo sapiens 106-110 31611911-11 2019 Moreover, we also noticed that the hallmark of epithelial-mesenchymal transition, ECM related biological processes and pathways were highly enriched in the MGMT unmethylated GBMs with the high-risk score, indicating that enhanced ECM activities could be involved in the TMZ-resistance of GBM. temozolomide 270-273 multimerin 1 Homo sapiens 82-85 31611911-11 2019 Moreover, we also noticed that the hallmark of epithelial-mesenchymal transition, ECM related biological processes and pathways were highly enriched in the MGMT unmethylated GBMs with the high-risk score, indicating that enhanced ECM activities could be involved in the TMZ-resistance of GBM. temozolomide 270-273 O-6-methylguanine-DNA methyltransferase Homo sapiens 156-160 31611911-11 2019 Moreover, we also noticed that the hallmark of epithelial-mesenchymal transition, ECM related biological processes and pathways were highly enriched in the MGMT unmethylated GBMs with the high-risk score, indicating that enhanced ECM activities could be involved in the TMZ-resistance of GBM. temozolomide 270-273 multimerin 1 Homo sapiens 230-233 31602000-6 2019 Further, BMP pathway activation confers relative resistance to radiation and temozolomide chemotherapy. temozolomide 77-89 bone morphogenetic protein 1 Homo sapiens 9-12 31686436-8 2019 After molecular and histological examinations, patients with anaplastic astrocytoma, isocitrate dehydrogenase (IDH)-mutant should be primary treated by standard brain radiotherapy and adjuvant temozolomide chemotherapy whereas those with anaplastic astrocytoma, NOS, and anaplastic astrocytoma, IDH-wildtype should be treated following the protocol for glioblastomas. temozolomide 193-205 isocitrate dehydrogenase (NADP(+)) 1 Homo sapiens 85-109 31686436-8 2019 After molecular and histological examinations, patients with anaplastic astrocytoma, isocitrate dehydrogenase (IDH)-mutant should be primary treated by standard brain radiotherapy and adjuvant temozolomide chemotherapy whereas those with anaplastic astrocytoma, NOS, and anaplastic astrocytoma, IDH-wildtype should be treated following the protocol for glioblastomas. temozolomide 193-205 isocitrate dehydrogenase (NADP(+)) 1 Homo sapiens 111-114 31561595-6 2019 Bioinformatics data have indicated that dopamine receptor (DRD) 2, DRD4, CD133 and Nestin were elevated in GBM clinical samples and correlated to Temozolomide (TMZ) resistance and increased aldehyde dehydrogenase (ALDH) activity (3.5-8.9%) as well as enhanced (2.1-2.4-fold) neurosphere formation efficiency in U87MG and D54MG GBM cell lines. temozolomide 160-163 dopamine receptor D4 Homo sapiens 67-71 31561595-7 2019 In addition, TMZ-resistant GSC phenotype was associated with up-regulated DRD4, Akt, mTOR, beta-catenin, CDK6, NF-kappaB and Erk1/2 expression. temozolomide 13-16 dopamine receptor D4 Homo sapiens 74-78 31561595-7 2019 In addition, TMZ-resistant GSC phenotype was associated with up-regulated DRD4, Akt, mTOR, beta-catenin, CDK6, NF-kappaB and Erk1/2 expression. temozolomide 13-16 AKT serine/threonine kinase 1 Homo sapiens 80-83 31561595-7 2019 In addition, TMZ-resistant GSC phenotype was associated with up-regulated DRD4, Akt, mTOR, beta-catenin, CDK6, NF-kappaB and Erk1/2 expression. temozolomide 13-16 mechanistic target of rapamycin kinase Homo sapiens 85-89 31561595-7 2019 In addition, TMZ-resistant GSC phenotype was associated with up-regulated DRD4, Akt, mTOR, beta-catenin, CDK6, NF-kappaB and Erk1/2 expression. temozolomide 13-16 catenin beta 1 Homo sapiens 91-103 31561595-7 2019 In addition, TMZ-resistant GSC phenotype was associated with up-regulated DRD4, Akt, mTOR, beta-catenin, CDK6, NF-kappaB and Erk1/2 expression. temozolomide 13-16 cyclin dependent kinase 6 Homo sapiens 105-109 31561595-7 2019 In addition, TMZ-resistant GSC phenotype was associated with up-regulated DRD4, Akt, mTOR, beta-catenin, CDK6, NF-kappaB and Erk1/2 expression. temozolomide 13-16 nuclear factor kappa B subunit 1 Homo sapiens 111-120 31561595-7 2019 In addition, TMZ-resistant GSC phenotype was associated with up-regulated DRD4, Akt, mTOR, beta-catenin, CDK6, NF-kappaB and Erk1/2 expression. temozolomide 13-16 mitogen-activated protein kinase 3 Homo sapiens 125-131 31561595-8 2019 LCC-09 alone, or combined with TMZ, suppressed the tumorigenic and stemness traits of TMZ-resistant GBM cells while concomitantly down-regulating DRD4, Akt, mTOR, beta-catenin, Erk1/2, NF-kappaB, and CDK6 expression. temozolomide 31-34 dopamine receptor D4 Homo sapiens 146-150 31561595-8 2019 LCC-09 alone, or combined with TMZ, suppressed the tumorigenic and stemness traits of TMZ-resistant GBM cells while concomitantly down-regulating DRD4, Akt, mTOR, beta-catenin, Erk1/2, NF-kappaB, and CDK6 expression. temozolomide 31-34 AKT serine/threonine kinase 1 Homo sapiens 152-155 31561595-8 2019 LCC-09 alone, or combined with TMZ, suppressed the tumorigenic and stemness traits of TMZ-resistant GBM cells while concomitantly down-regulating DRD4, Akt, mTOR, beta-catenin, Erk1/2, NF-kappaB, and CDK6 expression. temozolomide 31-34 mechanistic target of rapamycin kinase Homo sapiens 157-161 31561595-8 2019 LCC-09 alone, or combined with TMZ, suppressed the tumorigenic and stemness traits of TMZ-resistant GBM cells while concomitantly down-regulating DRD4, Akt, mTOR, beta-catenin, Erk1/2, NF-kappaB, and CDK6 expression. temozolomide 31-34 catenin beta 1 Homo sapiens 163-175 31750824-0 2019 [RAD51 promotes proliferation and migration of glioblastoma cells and decreases sensitivity of cells to temozolomide]. temozolomide 104-116 RAD51 recombinase Homo sapiens 1-6 31750824-1 2019 Objective To investigate the role of RAD51 in cell proliferation, migration and chemosensitivity to temozolomide (TMZ) using U251 glioma cell line, and to clarify the underlying molecular mechanism. temozolomide 100-112 RAD51 recombinase Homo sapiens 37-42 31750824-1 2019 Objective To investigate the role of RAD51 in cell proliferation, migration and chemosensitivity to temozolomide (TMZ) using U251 glioma cell line, and to clarify the underlying molecular mechanism. temozolomide 114-117 RAD51 recombinase Homo sapiens 37-42 31750824-7 2019 RAD51 enhanced the proliferation and migration ability of U251 glioma cells; knockdown of RAD51 enhanced the sensitivity of U251 glioma cells to temozolomide. temozolomide 145-157 RAD51 recombinase Homo sapiens 90-95 31750824-11 2019 RAD51 knockdown increases the sensitivity of glioma cells to temozolomide. temozolomide 61-73 RAD51 recombinase Homo sapiens 0-5 31818145-3 2019 We describe a case of CD10-positive PCNSL presenting with multiple posterior fossa enhancing lesions in an immunocompetent older woman with a history of breast cancer successfully treated by the RTOG 0227 protocol consisting of pre-irradiation chemotherapy with high-dose methotrexate, rituximab, and temozolomide for 6 cycles, followed by low-dose whole-brain radiation and post-irradiation temozolomide. temozolomide 301-313 membrane metalloendopeptidase Homo sapiens 22-26 31818145-3 2019 We describe a case of CD10-positive PCNSL presenting with multiple posterior fossa enhancing lesions in an immunocompetent older woman with a history of breast cancer successfully treated by the RTOG 0227 protocol consisting of pre-irradiation chemotherapy with high-dose methotrexate, rituximab, and temozolomide for 6 cycles, followed by low-dose whole-brain radiation and post-irradiation temozolomide. temozolomide 392-404 membrane metalloendopeptidase Homo sapiens 22-26 31798765-2 2019 O6-methyl-guanine DNA methyltransferase (MGMT) repairs DNA damage induced by TMZ; hence, elevated MGMT levels usually correlate with TMZ resistance. temozolomide 77-80 O-6-methylguanine-DNA methyltransferase Homo sapiens 0-39 31798765-2 2019 O6-methyl-guanine DNA methyltransferase (MGMT) repairs DNA damage induced by TMZ; hence, elevated MGMT levels usually correlate with TMZ resistance. temozolomide 77-80 O-6-methylguanine-DNA methyltransferase Homo sapiens 41-45 31798765-2 2019 O6-methyl-guanine DNA methyltransferase (MGMT) repairs DNA damage induced by TMZ; hence, elevated MGMT levels usually correlate with TMZ resistance. temozolomide 77-80 O-6-methylguanine-DNA methyltransferase Homo sapiens 98-102 31798765-2 2019 O6-methyl-guanine DNA methyltransferase (MGMT) repairs DNA damage induced by TMZ; hence, elevated MGMT levels usually correlate with TMZ resistance. temozolomide 133-136 O-6-methylguanine-DNA methyltransferase Homo sapiens 0-39 31798765-2 2019 O6-methyl-guanine DNA methyltransferase (MGMT) repairs DNA damage induced by TMZ; hence, elevated MGMT levels usually correlate with TMZ resistance. temozolomide 133-136 O-6-methylguanine-DNA methyltransferase Homo sapiens 41-45 31798765-2 2019 O6-methyl-guanine DNA methyltransferase (MGMT) repairs DNA damage induced by TMZ; hence, elevated MGMT levels usually correlate with TMZ resistance. temozolomide 133-136 O-6-methylguanine-DNA methyltransferase Homo sapiens 98-102 31798765-3 2019 MGMT promoter methylation is a key regulatory mechanism for MGMT expression and is important in overcoming TMZ therapy resistance. temozolomide 107-110 O-6-methylguanine-DNA methyltransferase Homo sapiens 0-4 31798765-3 2019 MGMT promoter methylation is a key regulatory mechanism for MGMT expression and is important in overcoming TMZ therapy resistance. temozolomide 107-110 O-6-methylguanine-DNA methyltransferase Homo sapiens 60-64 31798765-8 2019 Interestingly, this interaction is disrupted by TMZ exclusively in TMZ sensitive cells, suggesting that this MGMT regulatory pathway might be inactivated in TMZ resistant cells. temozolomide 48-51 O-6-methylguanine-DNA methyltransferase Homo sapiens 109-113 31798765-8 2019 Interestingly, this interaction is disrupted by TMZ exclusively in TMZ sensitive cells, suggesting that this MGMT regulatory pathway might be inactivated in TMZ resistant cells. temozolomide 67-70 O-6-methylguanine-DNA methyltransferase Homo sapiens 109-113 31798765-8 2019 Interestingly, this interaction is disrupted by TMZ exclusively in TMZ sensitive cells, suggesting that this MGMT regulatory pathway might be inactivated in TMZ resistant cells. temozolomide 67-70 O-6-methylguanine-DNA methyltransferase Homo sapiens 109-113