PMID-sentid Pub_year Sent_text compound_name comp_offset prot_official_name organism prot_offset 33887553-9 2021 In combination, sunitinib was able to overcome potential crizotinib-induced resistance through downregulation of ERK 1/2 activity and PDGFRss receptor expression; consequently, tumor growth was significantly decreased both in vitro and in vivo. Sunitinib 16-25 mitogen-activated protein kinase 3 Homo sapiens 113-120 34036385-0 2021 QPCT regulation by CTCF leads to sunitinib resistance in renal cell carcinoma by promoting angiogenesis. Sunitinib 33-42 glutaminyl-peptide cyclotransferase Homo sapiens 0-4 34036385-0 2021 QPCT regulation by CTCF leads to sunitinib resistance in renal cell carcinoma by promoting angiogenesis. Sunitinib 33-42 CCCTC-binding factor Homo sapiens 19-23 34036385-3 2021 It has previously been suggested that the protein glutaminyl-peptide cyclotransferase (QPCT) is closely related to sunitinib resistance in RCC. Sunitinib 115-124 glutaminyl-peptide cyclotransferase Homo sapiens 50-85 34036385-3 2021 It has previously been suggested that the protein glutaminyl-peptide cyclotransferase (QPCT) is closely related to sunitinib resistance in RCC. Sunitinib 115-124 glutaminyl-peptide cyclotransferase Homo sapiens 87-91 34036385-6 2021 On the whole, the data from the present study suggest that QPCT, CCCTC-binding factor (CTCF) and phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA) may be used as targets for predicting, reversing and treating sunitinib-resistant RCC. Sunitinib 239-248 glutaminyl-peptide cyclotransferase Homo sapiens 59-63 34036385-6 2021 On the whole, the data from the present study suggest that QPCT, CCCTC-binding factor (CTCF) and phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA) may be used as targets for predicting, reversing and treating sunitinib-resistant RCC. Sunitinib 239-248 phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha Homo sapiens 97-167 34036385-6 2021 On the whole, the data from the present study suggest that QPCT, CCCTC-binding factor (CTCF) and phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA) may be used as targets for predicting, reversing and treating sunitinib-resistant RCC. Sunitinib 239-248 phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha Homo sapiens 169-175 34057619-3 2021 Variants of the HT-29 line with complete or partial knockout of the PROM1 gene were equally sensitive to protein kinase inhibitors sorafenib and sunitinib. Sunitinib 145-154 prominin 1 Homo sapiens 68-73 33995090-0 2021 Sodium-Glucose CoTransporter-2 Inhibitor Empagliflozin Ameliorates Sunitinib-Induced Cardiac Dysfunction via Regulation of AMPK-mTOR Signaling Pathway-Mediated Autophagy. Sunitinib 67-76 solute carrier family 5 member 2 Homo sapiens 0-30 33893075-3 2021 MATERIALS AND METHODS: Parental and sunitinib-resistant 786-O cells were treated with GPER1 agonist G-1, and quantitative phosphoproteomics was performed. Sunitinib 36-45 G protein-coupled estrogen receptor 1 Homo sapiens 86-91 33893075-6 2021 Phosphoproteomic signatures, including phosphoinositide 3-kinase and protein kinase B (PI3K-AKT) as well as other pathways, were up-regulated in sunitinib-resistant cells but application of G-1 reversed this effect. Sunitinib 145-154 protein tyrosine kinase 2 beta Homo sapiens 69-85 33893075-6 2021 Phosphoproteomic signatures, including phosphoinositide 3-kinase and protein kinase B (PI3K-AKT) as well as other pathways, were up-regulated in sunitinib-resistant cells but application of G-1 reversed this effect. Sunitinib 145-154 AKT serine/threonine kinase 1 Homo sapiens 92-95 33893075-7 2021 Among phosphoprotein candidates, activating transcription factor 2 (ATF2) Thr69/71 phosphorylation was antagonistically regulated by sunitinib resistance and G-1. Sunitinib 133-142 activating transcription factor 2 Homo sapiens 33-66 33893075-7 2021 Among phosphoprotein candidates, activating transcription factor 2 (ATF2) Thr69/71 phosphorylation was antagonistically regulated by sunitinib resistance and G-1. Sunitinib 133-142 activating transcription factor 2 Homo sapiens 68-72 33893075-8 2021 CONCLUSION: Our results open up the possibility for managing RCC and sunitinib resistance by GPER1 agonist G-1 and its regulated pathways. Sunitinib 69-78 G protein-coupled estrogen receptor 1 Homo sapiens 93-98 33995090-0 2021 Sodium-Glucose CoTransporter-2 Inhibitor Empagliflozin Ameliorates Sunitinib-Induced Cardiac Dysfunction via Regulation of AMPK-mTOR Signaling Pathway-Mediated Autophagy. Sunitinib 67-76 protein kinase AMP-activated catalytic subunit alpha 2 Homo sapiens 123-127 33995090-0 2021 Sodium-Glucose CoTransporter-2 Inhibitor Empagliflozin Ameliorates Sunitinib-Induced Cardiac Dysfunction via Regulation of AMPK-mTOR Signaling Pathway-Mediated Autophagy. Sunitinib 67-76 mechanistic target of rapamycin kinase Homo sapiens 128-132 33995090-12 2021 Conclusion: EMPA could ameliorate SNT-induced cardiac dysfunction via regulating cardiomyocyte autophagy, which was mediated by the AMPK-mTOR signaling pathway. Sunitinib 34-37 protein kinase AMP-activated catalytic subunit alpha 2 Homo sapiens 132-136 33995090-12 2021 Conclusion: EMPA could ameliorate SNT-induced cardiac dysfunction via regulating cardiomyocyte autophagy, which was mediated by the AMPK-mTOR signaling pathway. Sunitinib 34-37 mechanistic target of rapamycin kinase Homo sapiens 137-141 33900546-4 2022 Here, we describe a hypotonic, thermosensitive gel-forming eye drop for effective delivery of sunitinib, a protein kinase inhibitor with activity against the neuroprotective targets dual leucine zipper kinase (DLK) and leucine zipper kinase (LZK), to enhance survival of RGCs after optic nerve injury. Sunitinib 94-103 mitogen-activated protein kinase kinase kinase 12 Homo sapiens 182-208 33906367-7 2021 Cobimetinib at nontoxic doses prevented tumor formation, inhibited tumor growth and enhanced efficacy of 5-fluorouracil, sorafenib and sunitinib via suppressing Raf/MEK/ERK, leading to MAPK pathway inhibition. Sunitinib 135-144 mitogen-activated protein kinase kinase 7 Homo sapiens 165-168 33906367-7 2021 Cobimetinib at nontoxic doses prevented tumor formation, inhibited tumor growth and enhanced efficacy of 5-fluorouracil, sorafenib and sunitinib via suppressing Raf/MEK/ERK, leading to MAPK pathway inhibition. Sunitinib 135-144 mitogen-activated protein kinase 1 Homo sapiens 169-172 33900546-4 2022 Here, we describe a hypotonic, thermosensitive gel-forming eye drop for effective delivery of sunitinib, a protein kinase inhibitor with activity against the neuroprotective targets dual leucine zipper kinase (DLK) and leucine zipper kinase (LZK), to enhance survival of RGCs after optic nerve injury. Sunitinib 94-103 mitogen-activated protein kinase kinase kinase 13 Homo sapiens 242-245 33900546-4 2022 Here, we describe a hypotonic, thermosensitive gel-forming eye drop for effective delivery of sunitinib, a protein kinase inhibitor with activity against the neuroprotective targets dual leucine zipper kinase (DLK) and leucine zipper kinase (LZK), to enhance survival of RGCs after optic nerve injury. Sunitinib 94-103 mitogen-activated protein kinase kinase kinase 12 Homo sapiens 210-213 33900546-4 2022 Here, we describe a hypotonic, thermosensitive gel-forming eye drop for effective delivery of sunitinib, a protein kinase inhibitor with activity against the neuroprotective targets dual leucine zipper kinase (DLK) and leucine zipper kinase (LZK), to enhance survival of RGCs after optic nerve injury. Sunitinib 94-103 mitogen-activated protein kinase kinase kinase 13 Homo sapiens 187-208 33854032-0 2021 Correction: ZHX2 drives cell growth and migration via activating MEK/ERK signal and induces Sunitinib resistance by regulating the autophagy in clear cell Renal Cell Carcinoma. Sunitinib 92-101 zinc fingers and homeoboxes 2 Homo sapiens 12-16 33863648-1 2021 BACKGROUND: Limited data exist on the clinical effectiveness of second-line (2L) vascular endothelial growth factor (receptor) targeted inhibitor (VEGF(R)i) sunitinib after first-line (1L) immuno-oncology (IO) therapy for patients with metastatic renal cell carcinoma (mRCC) in real-world settings. Sunitinib 157-166 vascular endothelial growth factor A Homo sapiens 81-145 33936816-0 2021 Sunitinib Combined with Th1 Cytokines Potentiates Apoptosis in Human Breast Cancer Cells and Suppresses Tumor Growth in a Murine Model of HER-2pos Breast Cancer. Sunitinib 0-9 erb-b2 receptor tyrosine kinase 2 Mus musculus 138-143 33936816-6 2021 Interestingly, sunitinib was found to have a profoundly suppressive effect of T cell"s capacity to secrete IFN-gamma, indicating that in vivo use of this drug may hinder robust Th1 responses. Sunitinib 15-24 interferon gamma Homo sapiens 107-116 33647840-4 2021 Compounds 1 and 4 inhibited VEGFR-2 kinase with activity better than or equal to that of sunitinib. Sunitinib 89-98 kinase insert domain receptor Homo sapiens 28-35 33732370-9 2021 Therefore, it is proposed that PDGFR inhibitors, including sunitinib and ponatinib, should be applied effectively to treat ER-alpha+ breast cancer. Sunitinib 59-68 platelet derived growth factor receptor beta Homo sapiens 31-36 33732370-9 2021 Therefore, it is proposed that PDGFR inhibitors, including sunitinib and ponatinib, should be applied effectively to treat ER-alpha+ breast cancer. Sunitinib 59-68 estrogen receptor 1 Homo sapiens 123-131 33842334-7 2021 Application of AhR inhibitor DMF could efficiently suppress distant metastasis of renal cancer cells, and improve anticancer effects of sorafenib (Sor)/sunitinib (Sun), which described a promising therapeutic strategy for clinical renal cancer. Sunitinib 152-161 aryl hydrocarbon receptor Homo sapiens 15-18 33842334-7 2021 Application of AhR inhibitor DMF could efficiently suppress distant metastasis of renal cancer cells, and improve anticancer effects of sorafenib (Sor)/sunitinib (Sun), which described a promising therapeutic strategy for clinical renal cancer. Sunitinib 163-166 aryl hydrocarbon receptor Homo sapiens 15-18 33841146-9 2021 Furthermore, western blot analysis revealed that the expressions levels of phosphorylated IRS-1, Pellino-1, AKT and eNOS were significantly attenuated by sunitinib in rat mesenteric artery tissues and in the sunitinib-treated primary cultured mesenteric artery endothelial cells. Sunitinib 154-163 insulin receptor substrate 1 Rattus norvegicus 90-95 33841146-9 2021 Furthermore, western blot analysis revealed that the expressions levels of phosphorylated IRS-1, Pellino-1, AKT and eNOS were significantly attenuated by sunitinib in rat mesenteric artery tissues and in the sunitinib-treated primary cultured mesenteric artery endothelial cells. Sunitinib 154-163 pellino E3 ubiquitin protein ligase 1 Rattus norvegicus 97-106 33841146-9 2021 Furthermore, western blot analysis revealed that the expressions levels of phosphorylated IRS-1, Pellino-1, AKT and eNOS were significantly attenuated by sunitinib in rat mesenteric artery tissues and in the sunitinib-treated primary cultured mesenteric artery endothelial cells. Sunitinib 154-163 AKT serine/threonine kinase 1 Rattus norvegicus 108-111 33841146-9 2021 Furthermore, western blot analysis revealed that the expressions levels of phosphorylated IRS-1, Pellino-1, AKT and eNOS were significantly attenuated by sunitinib in rat mesenteric artery tissues and in the sunitinib-treated primary cultured mesenteric artery endothelial cells. Sunitinib 154-163 nitric oxide synthase 3 Rattus norvegicus 116-120 33841146-9 2021 Furthermore, western blot analysis revealed that the expressions levels of phosphorylated IRS-1, Pellino-1, AKT and eNOS were significantly attenuated by sunitinib in rat mesenteric artery tissues and in the sunitinib-treated primary cultured mesenteric artery endothelial cells. Sunitinib 208-217 insulin receptor substrate 1 Rattus norvegicus 90-95 33841146-9 2021 Furthermore, western blot analysis revealed that the expressions levels of phosphorylated IRS-1, Pellino-1, AKT and eNOS were significantly attenuated by sunitinib in rat mesenteric artery tissues and in the sunitinib-treated primary cultured mesenteric artery endothelial cells. Sunitinib 208-217 nitric oxide synthase 3 Rattus norvegicus 116-120 33841146-11 2021 Moreover, sunitinib-induced decrease in the expression levels of phosphorylated AKT and eNOS was further reduced by knocking down of Pellino-1 in MAECs. Sunitinib 10-19 AKT serine/threonine kinase 1 Rattus norvegicus 80-83 33841146-11 2021 Moreover, sunitinib-induced decrease in the expression levels of phosphorylated AKT and eNOS was further reduced by knocking down of Pellino-1 in MAECs. Sunitinib 10-19 nitric oxide synthase 3 Rattus norvegicus 88-92 33841146-11 2021 Moreover, sunitinib-induced decrease in the expression levels of phosphorylated AKT and eNOS was further reduced by knocking down of Pellino-1 in MAECs. Sunitinib 10-19 pellino E3 ubiquitin protein ligase 1 Rattus norvegicus 133-142 33841146-12 2021 Our results suggest that sunitinib causes vascular dysfunction and hypertension, which are associated with insulin resistance- and Pellino-1-mediated inhibition of AKT/eNOS/NO signaling. Sunitinib 25-34 pellino E3 ubiquitin protein ligase 1 Rattus norvegicus 131-140 33841146-12 2021 Our results suggest that sunitinib causes vascular dysfunction and hypertension, which are associated with insulin resistance- and Pellino-1-mediated inhibition of AKT/eNOS/NO signaling. Sunitinib 25-34 AKT serine/threonine kinase 1 Rattus norvegicus 164-167 33841146-12 2021 Our results suggest that sunitinib causes vascular dysfunction and hypertension, which are associated with insulin resistance- and Pellino-1-mediated inhibition of AKT/eNOS/NO signaling. Sunitinib 25-34 nitric oxide synthase 3 Rattus norvegicus 168-172 33976736-11 2021 C2 and C3 were corresponding to a better survival outcome and presented a low drug sensitivity to sorafenib and sunitinib. Sunitinib 112-121 complement C2 Homo sapiens 0-9 32530390-4 2021 The cells were treated with an inhibitor of either ACK1 (dasatinib or sunitinib) or AKT (MK-2206 or GDC-0068), and the optimal concentrations of the two yielding synergistic tumor-killing effects were determined by applying the Chou-Talalay equation for drug combinations. Sunitinib 70-79 tyrosine kinase non receptor 2 Homo sapiens 51-55 31331862-10 2021 CONCLUSIONS: Treatment with further TKIs or mTOR inhibitors for mRCC patients primarily refractory to first-line sunitinib in the observed time period achieved very minimal benefit, suggesting avoiding TKI rechallenge and possibly preferring alternative strategies, such as immune checkpoint inhibitors, after PD to a treatment line including a TKI in this setting. Sunitinib 113-122 mechanistic target of rapamycin kinase Homo sapiens 44-48 33732370-4 2021 To block the PDGF-BB signaling pathway, PDGFR inhibitors (sunitinib or ponatinib) were employed. Sunitinib 58-67 platelet derived growth factor receptor beta Homo sapiens 40-45 33723657-0 2021 Is there association between clinically relevant toxicities of pazopanib and sunitinib with the use of weak CYP3A4 and P-gp inhibitors? Sunitinib 77-86 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 108-114 33723657-0 2021 Is there association between clinically relevant toxicities of pazopanib and sunitinib with the use of weak CYP3A4 and P-gp inhibitors? Sunitinib 77-86 phosphoglycolate phosphatase Homo sapiens 119-123 33135264-9 2021 Thus, sunitinib reduces the expression of KRT6A and SERPINB1 by inhibiting the ERK1/2 and p38 MAPK signaling pathways in the skin model. Sunitinib 6-15 keratin 6A Homo sapiens 42-47 33722287-11 2021 Sunitinib (SNT)-induced upregulation of glial fibrillary acidic protein (GFAP) (astrocyte) and ionized calcium-binding adaptor molecule 1 (Iba-1) (microglia) in the ipsilateral L4-5 dorsal and ventral horn relative to the contralateral side. Sunitinib 0-9 glial fibrillary acidic protein Homo sapiens 40-71 33722287-11 2021 Sunitinib (SNT)-induced upregulation of glial fibrillary acidic protein (GFAP) (astrocyte) and ionized calcium-binding adaptor molecule 1 (Iba-1) (microglia) in the ipsilateral L4-5 dorsal and ventral horn relative to the contralateral side. Sunitinib 0-9 glial fibrillary acidic protein Homo sapiens 73-77 33722287-11 2021 Sunitinib (SNT)-induced upregulation of glial fibrillary acidic protein (GFAP) (astrocyte) and ionized calcium-binding adaptor molecule 1 (Iba-1) (microglia) in the ipsilateral L4-5 dorsal and ventral horn relative to the contralateral side. Sunitinib 0-9 allograft inflammatory factor 1 Homo sapiens 139-144 33859753-10 2021 Moreover, PRMT1 inhibition by DCPT1061 not only inhibited tumor growth but also sensitized ccRCC to sunitinib treatment in vivo by attenuating sunitinib-induced upregulation of LCN2-AKT-RB signaling. Sunitinib 100-109 protein arginine methyltransferase 1 Homo sapiens 10-15 33859753-10 2021 Moreover, PRMT1 inhibition by DCPT1061 not only inhibited tumor growth but also sensitized ccRCC to sunitinib treatment in vivo by attenuating sunitinib-induced upregulation of LCN2-AKT-RB signaling. Sunitinib 143-152 protein arginine methyltransferase 1 Homo sapiens 10-15 33859753-10 2021 Moreover, PRMT1 inhibition by DCPT1061 not only inhibited tumor growth but also sensitized ccRCC to sunitinib treatment in vivo by attenuating sunitinib-induced upregulation of LCN2-AKT-RB signaling. Sunitinib 143-152 lipocalin 2 Homo sapiens 177-181 33707524-9 2021 In parallel, in rats exposed to sunitinib, glomerular thrombomodulin was upregulated in a dose-dependent manner, and the upregulation of glomerular thrombomodulin preceded the onset of histopathological changes. Sunitinib 32-41 thrombomodulin Rattus norvegicus 54-68 33707524-9 2021 In parallel, in rats exposed to sunitinib, glomerular thrombomodulin was upregulated in a dose-dependent manner, and the upregulation of glomerular thrombomodulin preceded the onset of histopathological changes. Sunitinib 32-41 thrombomodulin Rattus norvegicus 148-162 33707524-10 2021 Selective ETAR blockade, but not dual ETA/BR blockade, normalised the sunitinib-induced increase in thrombomodulin expression and albuminuria. Sunitinib 70-79 endothelin receptor type A Homo sapiens 10-14 33707524-10 2021 Selective ETAR blockade, but not dual ETA/BR blockade, normalised the sunitinib-induced increase in thrombomodulin expression and albuminuria. Sunitinib 70-79 thrombomodulin Rattus norvegicus 100-114 33307872-6 2021 Sunitinib and regorafenib are inhibitors of multiple tyrosine kinases, including KIT, PDGFRalpha, PDGFRbeta, and VEGFRs, and are approved for the management of imatinib- and imatinib/sunitinib-refractory GIST patients, respectively. Sunitinib 0-9 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 81-84 33307872-6 2021 Sunitinib and regorafenib are inhibitors of multiple tyrosine kinases, including KIT, PDGFRalpha, PDGFRbeta, and VEGFRs, and are approved for the management of imatinib- and imatinib/sunitinib-refractory GIST patients, respectively. Sunitinib 0-9 platelet derived growth factor receptor alpha Homo sapiens 86-96 33307872-6 2021 Sunitinib and regorafenib are inhibitors of multiple tyrosine kinases, including KIT, PDGFRalpha, PDGFRbeta, and VEGFRs, and are approved for the management of imatinib- and imatinib/sunitinib-refractory GIST patients, respectively. Sunitinib 0-9 platelet derived growth factor receptor beta Homo sapiens 98-107 33135264-0 2021 Sunitinib decreases the expression of KRT6A and SERPINB1 in 3D human epidermal models. Sunitinib 0-9 keratin 6A Homo sapiens 38-43 33135264-0 2021 Sunitinib decreases the expression of KRT6A and SERPINB1 in 3D human epidermal models. Sunitinib 0-9 serpin family B member 1 Homo sapiens 48-56 33135264-5 2021 Sunitinib treatment significantly decreased the expression of SERPINB1 and KRT6A in the immunohistochemical staining of the 3D epidermal model. Sunitinib 0-9 serpin family B member 1 Homo sapiens 62-70 33135264-5 2021 Sunitinib treatment significantly decreased the expression of SERPINB1 and KRT6A in the immunohistochemical staining of the 3D epidermal model. Sunitinib 0-9 keratin 6A Homo sapiens 75-80 33135264-6 2021 In PSVK1 cells, but not in normal human epidermal keratinocyte cells, both of which are human normal keratinocyte cell lines, sunitinib decreased the expression of KRT6A with a concomitant decrease in levels of phosphorylated extracellular signal-regulated kinases (ERK)1/2 and phosphorylated p38 mitogen-activated protein kinase (MAPK). Sunitinib 126-135 keratin 6A Homo sapiens 164-169 33135264-6 2021 In PSVK1 cells, but not in normal human epidermal keratinocyte cells, both of which are human normal keratinocyte cell lines, sunitinib decreased the expression of KRT6A with a concomitant decrease in levels of phosphorylated extracellular signal-regulated kinases (ERK)1/2 and phosphorylated p38 mitogen-activated protein kinase (MAPK). Sunitinib 126-135 mitogen-activated protein kinase 3 Homo sapiens 266-273 33135264-6 2021 In PSVK1 cells, but not in normal human epidermal keratinocyte cells, both of which are human normal keratinocyte cell lines, sunitinib decreased the expression of KRT6A with a concomitant decrease in levels of phosphorylated extracellular signal-regulated kinases (ERK)1/2 and phosphorylated p38 mitogen-activated protein kinase (MAPK). Sunitinib 126-135 mitogen-activated protein kinase 14 Homo sapiens 293-329 33135264-8 2021 Sunitinib-induced decrease in KRT6A expression was suppressed by the inhibition of glycogen synthase kinase-3beta by enhancing ERK1/2 and p38 MAPK phosphorylation. Sunitinib 0-9 keratin 6A Homo sapiens 30-35 33135264-9 2021 Thus, sunitinib reduces the expression of KRT6A and SERPINB1 by inhibiting the ERK1/2 and p38 MAPK signaling pathways in the skin model. Sunitinib 6-15 serpin family B member 1 Homo sapiens 52-60 33135264-8 2021 Sunitinib-induced decrease in KRT6A expression was suppressed by the inhibition of glycogen synthase kinase-3beta by enhancing ERK1/2 and p38 MAPK phosphorylation. Sunitinib 0-9 glycogen synthase kinase 3 beta Homo sapiens 83-113 33135264-8 2021 Sunitinib-induced decrease in KRT6A expression was suppressed by the inhibition of glycogen synthase kinase-3beta by enhancing ERK1/2 and p38 MAPK phosphorylation. Sunitinib 0-9 mitogen-activated protein kinase 3 Homo sapiens 127-133 33135264-9 2021 Thus, sunitinib reduces the expression of KRT6A and SERPINB1 by inhibiting the ERK1/2 and p38 MAPK signaling pathways in the skin model. Sunitinib 6-15 mitogen-activated protein kinase 3 Homo sapiens 79-85 33593885-14 2021 CONCLUSIONS: In patients with metastatic non-clear cell RCC, we identified several poor prognosis angiokines and immunomodulatory chemokines during treatment with sunitinib or everolimus, particularly OPN. Sunitinib 163-172 secreted phosphoprotein 1 Homo sapiens 201-204 33455080-0 2021 Dysregulation of the Sirt5/IDH2 axis contributes to sunitinib resistance in human renal cancer cells. Sunitinib 52-61 sirtuin 5 Homo sapiens 21-26 33455080-0 2021 Dysregulation of the Sirt5/IDH2 axis contributes to sunitinib resistance in human renal cancer cells. Sunitinib 52-61 isocitrate dehydrogenase (NADP(+)) 2 Homo sapiens 27-31 33455080-1 2021 Sunitinib (Sun), a tyrosine kinase inhibitor of vascular endothelial growth factor receptor (VEGFR), is the standard first-line treatment against advanced clear cell renal cell carcinoma (ccRCC), but resistance to therapy is inevitable. Sunitinib 0-9 kinase insert domain receptor Homo sapiens 48-91 33455080-1 2021 Sunitinib (Sun), a tyrosine kinase inhibitor of vascular endothelial growth factor receptor (VEGFR), is the standard first-line treatment against advanced clear cell renal cell carcinoma (ccRCC), but resistance to therapy is inevitable. Sunitinib 0-9 kinase insert domain receptor Homo sapiens 93-98 33455080-1 2021 Sunitinib (Sun), a tyrosine kinase inhibitor of vascular endothelial growth factor receptor (VEGFR), is the standard first-line treatment against advanced clear cell renal cell carcinoma (ccRCC), but resistance to therapy is inevitable. Sunitinib 0-3 kinase insert domain receptor Homo sapiens 48-91 33455080-1 2021 Sunitinib (Sun), a tyrosine kinase inhibitor of vascular endothelial growth factor receptor (VEGFR), is the standard first-line treatment against advanced clear cell renal cell carcinoma (ccRCC), but resistance to therapy is inevitable. Sunitinib 0-3 kinase insert domain receptor Homo sapiens 93-98 33717062-10 2021 Results: Along with its anti-proliferative and direct pro-apoptotic effect on sarcoma cell lines, sunitinib prompted PD-L1 upregulation on sarcoma cells. Sunitinib 98-107 CD274 molecule Homo sapiens 117-122 33717062-11 2021 Interestingly, sunitinib-treated sarcoma cells drive DCs to full maturation and increase their capacity to induce sarcoma-reactive T cells to produce IFN-gamma. Sunitinib 15-24 interferon gamma Sus scrofa 150-159 33717062-15 2021 Indeed, PD-1 blockade by nivolumab synergized with sunitinib in inducing IFN-gamma-producing effector T cells. Sunitinib 51-60 interferon gamma Sus scrofa 73-82 33717062-16 2021 Conclusions: Taken together, our in vitro data indicate that the treatment of sarcoma cells with sunitinib can exert significant changes on immune cell subsets toward immune activation, leading to DC-based cross-priming of IFN-gamma-producing effector T cells and reduced Treg induction. Sunitinib 97-106 interferon gamma Sus scrofa 223-232 33592176-17 2021 INTERPRETATION: Cabozantinib treatment resulted in significantly longer PFS compared with sunitinib in patients with metastatic PRCC. Sunitinib 90-99 proline rich mitotic checkpoint control factor Homo sapiens 128-132 33078449-7 2021 The docking study of the most potent compound against VEGFR-2 with the best-scored conformations displayed a binding affinity (-9.5 kcal/mol) comparable with the drug sunitinib (-9.9 kcal/mol) and exhibited that tighter interactions at the active adenosine triphosphate site might be responsible for anticancer potency. Sunitinib 167-176 kinase insert domain receptor Homo sapiens 54-61 33547392-8 2021 High Plk1 expression conferred a resistant phenotype of ccRCC to targeted therapeutics such as sunitinib, in vitro, in vivo, and in metastatic ccRCC patients. Sunitinib 95-104 polo like kinase 1 Homo sapiens 5-9 33597889-7 2021 In vivo studies further confirmed that crizotinib and sunitinib decreased mitochondrial membrane potential and activated apoptosis-associated proteins (cleaved-PARP, cleaved caspase3, cytochrome c, Bcl2 and Bax). Sunitinib 54-63 collagen type XI alpha 2 chain Homo sapiens 160-164 33634984-0 2021 Epigenome screening highlights that JMJD6 confers an epigenetic vulnerability and mediates sunitinib sensitivity in renal cell carcinoma. Sunitinib 91-100 jumonji domain containing 6, arginine demethylase and lysine hydroxylase Homo sapiens 36-41 33634984-8 2021 Given the probable overlapped crosstalk between JMJD6 signature and tyrosine kinase inhibitors downstream targets, targeting JMJD6 sensitized RCC to sunitinib and was synergistic when they were combined together. Sunitinib 149-158 jumonji domain containing 6, arginine demethylase and lysine hydroxylase Homo sapiens 125-130 33597889-0 2021 Crizotinib and Sunitinib Induce Hepatotoxicity and Mitochondrial Apoptosis in L02 Cells via ROS and Nrf2 Signaling Pathway. Sunitinib 15-24 NFE2 like bZIP transcription factor 2 Homo sapiens 100-104 33598481-12 2020 Both sunitinib and imatinib pre-treatment increased Angiotensin II-induced intracellular Ca2+ mobilization, with only sunitinib resulting in a significant effect and also causing increased CaMKII activation via oxidation. Sunitinib 5-14 angiotensinogen Rattus norvegicus 52-66 33597889-7 2021 In vivo studies further confirmed that crizotinib and sunitinib decreased mitochondrial membrane potential and activated apoptosis-associated proteins (cleaved-PARP, cleaved caspase3, cytochrome c, Bcl2 and Bax). Sunitinib 54-63 caspase 3 Homo sapiens 174-182 33598481-16 2020 Sunitinib caused a significant increase in Angiotensin II-induced intracellular Ca2+ mobilization and both TKIs caused increased mitochondrial superoxide production. Sunitinib 0-9 angiotensinogen Rattus norvegicus 43-57 33597889-7 2021 In vivo studies further confirmed that crizotinib and sunitinib decreased mitochondrial membrane potential and activated apoptosis-associated proteins (cleaved-PARP, cleaved caspase3, cytochrome c, Bcl2 and Bax). Sunitinib 54-63 cytochrome c, somatic Homo sapiens 184-196 33597889-7 2021 In vivo studies further confirmed that crizotinib and sunitinib decreased mitochondrial membrane potential and activated apoptosis-associated proteins (cleaved-PARP, cleaved caspase3, cytochrome c, Bcl2 and Bax). Sunitinib 54-63 BCL2 apoptosis regulator Homo sapiens 198-202 33597889-7 2021 In vivo studies further confirmed that crizotinib and sunitinib decreased mitochondrial membrane potential and activated apoptosis-associated proteins (cleaved-PARP, cleaved caspase3, cytochrome c, Bcl2 and Bax). Sunitinib 54-63 BCL2 associated X, apoptosis regulator Homo sapiens 207-210 33597889-8 2021 Furthermore, mechanistic investigations demonstrated that crizotinib and sunitinib accumulated ROS and inhibited Nrf2 signaling, and that ROS scavenger NAC and Nrf2 agonist tBHQ alleviated the extent of cell damage and the mitochondrial apoptosis during crizotinib- and sunitinib-induced hepatotoxicity in L02 cells. Sunitinib 73-82 NFE2 like bZIP transcription factor 2 Homo sapiens 113-117 33597889-8 2021 Furthermore, mechanistic investigations demonstrated that crizotinib and sunitinib accumulated ROS and inhibited Nrf2 signaling, and that ROS scavenger NAC and Nrf2 agonist tBHQ alleviated the extent of cell damage and the mitochondrial apoptosis during crizotinib- and sunitinib-induced hepatotoxicity in L02 cells. Sunitinib 270-279 X-linked Kx blood group Homo sapiens 152-155 33597889-8 2021 Furthermore, mechanistic investigations demonstrated that crizotinib and sunitinib accumulated ROS and inhibited Nrf2 signaling, and that ROS scavenger NAC and Nrf2 agonist tBHQ alleviated the extent of cell damage and the mitochondrial apoptosis during crizotinib- and sunitinib-induced hepatotoxicity in L02 cells. Sunitinib 270-279 NFE2 like bZIP transcription factor 2 Homo sapiens 160-164 33657773-0 2021 [Gas-Producing Splenic Abscess after Spleen Metastasis of Left Renal Cell Carcinoma Spleen Metastasis Successfully Treated with Sunitinib]. Sunitinib 128-137 galactosamine (N-acetyl)-6-sulfatase Homo sapiens 1-4 33597889-9 2021 Collectively, these findings indicated that NAC and tBHQ play the crucial roles in crizotinib- and sunitinib-induced mitochondrial apoptosis via the regulation of oxidative stress. Sunitinib 99-108 X-linked Kx blood group Homo sapiens 44-47 33376527-10 2021 rs191188930 on platelet-derived growth factor receptor (PDGFR) alpha was associated with numerous TKI drugs, including sunitinib, pazopanib, sorafenib, dasatinib and nilotinib. Sunitinib 119-128 platelet derived growth factor receptor alpha Homo sapiens 56-68 33410111-0 2021 Guanabenz Sensitizes Glioblastoma Cells to Sunitinib by Inhibiting GADD34-Mediated Autophagic Signaling. Sunitinib 43-52 protein phosphatase 1 regulatory subunit 15A Homo sapiens 67-73 33410111-10 2021 Guanabenz, an alpha2-selective adrenergic agonist and GADD34 functional inhibitor, was identified to enhance the efficacy of sunitinib by targeting GADD34-induced protective autophagy in glioblastoma cells, TMZ-resistant cells, hypoxic cultured cells, sphere-forming cells, and colony formation abilities. Sunitinib 125-134 glycoprotein hormone subunit alpha 2 Homo sapiens 14-20 33410111-8 2021 Growth arrest and DNA damage-inducible protein (GADD) 34 was identified as a candidate involved in sunitinib-promoted autophagy through activating p38-mitogen-activated protein kinase (MAPK) signaling. Sunitinib 99-108 protein phosphatase 1 regulatory subunit 15A Homo sapiens 0-56 33410111-10 2021 Guanabenz, an alpha2-selective adrenergic agonist and GADD34 functional inhibitor, was identified to enhance the efficacy of sunitinib by targeting GADD34-induced protective autophagy in glioblastoma cells, TMZ-resistant cells, hypoxic cultured cells, sphere-forming cells, and colony formation abilities. Sunitinib 125-134 protein phosphatase 1 regulatory subunit 15A Homo sapiens 54-60 33410111-10 2021 Guanabenz, an alpha2-selective adrenergic agonist and GADD34 functional inhibitor, was identified to enhance the efficacy of sunitinib by targeting GADD34-induced protective autophagy in glioblastoma cells, TMZ-resistant cells, hypoxic cultured cells, sphere-forming cells, and colony formation abilities. Sunitinib 125-134 protein phosphatase 1 regulatory subunit 15A Homo sapiens 148-154 33410111-12 2021 Taken together, the sunitinib therapy combined with guanabenz in the inhibition of GADD34-enhanced protective autophagy may provide a new therapeutic strategy for glioblastoma. Sunitinib 20-29 protein phosphatase 1, regulatory subunit 15A Mus musculus 83-89 33410111-8 2021 Growth arrest and DNA damage-inducible protein (GADD) 34 was identified as a candidate involved in sunitinib-promoted autophagy through activating p38-mitogen-activated protein kinase (MAPK) signaling. Sunitinib 99-108 mitogen-activated protein kinase 14 Homo sapiens 147-183 33410111-8 2021 Growth arrest and DNA damage-inducible protein (GADD) 34 was identified as a candidate involved in sunitinib-promoted autophagy through activating p38-mitogen-activated protein kinase (MAPK) signaling. Sunitinib 99-108 mitogen-activated protein kinase 14 Homo sapiens 185-189 33410111-9 2021 Higher GADD34 levels predicted poor survival of glioblastoma patients and induced autophagy formation in desensitizing sunitinib cytotoxicity. Sunitinib 119-128 protein phosphatase 1 regulatory subunit 15A Homo sapiens 7-13 33309571-11 2021 Among individual VEGFR-TKIs, Sunitinib (OR = 3.31, 95% CI 2.34-4.69) and Regorafenib (OR = 2.92, 95% CI 1.50-5.71) were associated with higher risk of hemorrhagic events in comparison to placebo. Sunitinib 29-38 kinase insert domain receptor Homo sapiens 17-22 33170322-1 2021 The objective of present work is to evaluate possible interactions among four clinically-used vascular epidermal growth factor receptor (VEGFR)-tyrosine kinase inhibitors (TKIs), including apatinib, cabozantinib, sorafenib, and sunitinib, with epidermal growth factor receptor (EGFR)-TKI gefitinib. Sunitinib 228-237 kinase insert domain receptor Homo sapiens 94-135 33170322-1 2021 The objective of present work is to evaluate possible interactions among four clinically-used vascular epidermal growth factor receptor (VEGFR)-tyrosine kinase inhibitors (TKIs), including apatinib, cabozantinib, sorafenib, and sunitinib, with epidermal growth factor receptor (EGFR)-TKI gefitinib. Sunitinib 228-237 kinase insert domain receptor Homo sapiens 137-142 33170322-1 2021 The objective of present work is to evaluate possible interactions among four clinically-used vascular epidermal growth factor receptor (VEGFR)-tyrosine kinase inhibitors (TKIs), including apatinib, cabozantinib, sorafenib, and sunitinib, with epidermal growth factor receptor (EGFR)-TKI gefitinib. Sunitinib 228-237 epidermal growth factor receptor Homo sapiens 103-135 33191180-0 2021 Sunitinib malate inhibits intestinal tumor development in male ApcMin/+ mice by down-regulating inflammation-related factors with suppressing beta-cateinin/c-Myc pathway and re-balancing Bcl-6 and Caspase-3. Sunitinib 0-16 B cell leukemia/lymphoma 6 Mus musculus 187-192 33191180-0 2021 Sunitinib malate inhibits intestinal tumor development in male ApcMin/+ mice by down-regulating inflammation-related factors with suppressing beta-cateinin/c-Myc pathway and re-balancing Bcl-6 and Caspase-3. Sunitinib 0-16 caspase 3 Mus musculus 197-206 33191180-9 2021 These results suggest that sunitinib malate can significantly improve the survival status and inhibit intestinal tumor development in male ApcMin/+ mice, through inhibiting inflammation-related factors, while suppressing beta-cateinin/c-Myc pathway and re-balancing protein levels of Bcl-6 and Caspase-3. Sunitinib 27-43 B cell leukemia/lymphoma 6 Mus musculus 284-289 33191180-9 2021 These results suggest that sunitinib malate can significantly improve the survival status and inhibit intestinal tumor development in male ApcMin/+ mice, through inhibiting inflammation-related factors, while suppressing beta-cateinin/c-Myc pathway and re-balancing protein levels of Bcl-6 and Caspase-3. Sunitinib 27-43 caspase 3 Mus musculus 294-303 33179075-6 2021 The results of the present study demonstrated that sunitinib reduced PARP1 expression and proliferation of melanoma cells. Sunitinib 51-60 poly(ADP-ribose) polymerase 1 Homo sapiens 69-74 33179075-7 2021 Notably, one of the PARPis, veliparib, reversed the inhibitory effect of sunitinib on PARP1 expression and proliferation, indicating that inhibition of PARP1 enzyme activity by PARPi may be different from the inhibition of PARP1 expression in melanoma cell biological function. Sunitinib 73-82 poly(ADP-ribose) polymerase 1 Homo sapiens 86-91 33179075-7 2021 Notably, one of the PARPis, veliparib, reversed the inhibitory effect of sunitinib on PARP1 expression and proliferation, indicating that inhibition of PARP1 enzyme activity by PARPi may be different from the inhibition of PARP1 expression in melanoma cell biological function. Sunitinib 73-82 poly(ADP-ribose) polymerase 1 Homo sapiens 152-157 33179075-7 2021 Notably, one of the PARPis, veliparib, reversed the inhibitory effect of sunitinib on PARP1 expression and proliferation, indicating that inhibition of PARP1 enzyme activity by PARPi may be different from the inhibition of PARP1 expression in melanoma cell biological function. Sunitinib 73-82 poly(ADP-ribose) polymerase 1 Homo sapiens 152-157 33374949-6 2020 The established sunitinib-resistant RCC cell line demonstrated significantly desuppressed protein kinase B (Akt) and mesenchymal-to-epithelial transition (MET) phosphorylation compared with the control RCC cell line under sunitinib exposure. Sunitinib 16-25 thymoma viral proto-oncogene 1 Mus musculus 108-111 33374949-8 2020 Particularly, glutamine transporter (SLC1A5) expression was significantly increased in sunitinib-resistant RCC cells compared with control cells. Sunitinib 87-96 solute carrier family 1 (neutral amino acid transporter), member 5 Mus musculus 37-43 33145941-0 2020 TFE3-PD-L1 axis is pivotal for sunitinib resistance in clear cell renal cell carcinoma. Sunitinib 31-40 transcription factor binding to IGHM enhancer 3 Homo sapiens 0-4 32947227-0 2020 Novel 2-indolinone thiazole hybrids as sunitinib analogues: Design, synthesis, and potent VEGFR-2 inhibition with potential anti-renal cancer activity. Sunitinib 39-48 kinase insert domain receptor Homo sapiens 90-97 32947227-1 2020 Novel 2-indolinone thiazole hybrids were designed and synthesized as VEGFR-2 inhibitors based on sunitinib, an FDA-approved anticancer drug. Sunitinib 97-106 kinase insert domain receptor Homo sapiens 69-76 32947227-4 2020 All tested compounds exhibited a potent submicromolar inhibition of VEGFR-2 kinase with IC50 values ranging from 0.067 to 0.422 muM, relative to sunitinib reference drug (IC50 = 0.075 +- 0.002 muM). Sunitinib 145-154 kinase insert domain receptor Homo sapiens 68-75 32947227-6 2020 Compound 13b stood out as the most potent against VEGFR-2 showing IC50 value of 0.067 +- 0.002 muM, lower than that of sunitinib. Sunitinib 119-128 kinase insert domain receptor Homo sapiens 50-57 33038552-6 2020 Moreover, compounds 5a, 8c, 9d, 12c were equipotent to sunitinib against VEGFR-2 kinase. Sunitinib 55-64 kinase insert domain receptor Homo sapiens 73-80 33022096-7 2020 RESULTS: Across all utility combinations tested, Q-TWiST was found to be longer with cabozantinib versus sunitinib (range of differences, +24 days to +137 days). Sunitinib 105-114 twist family bHLH transcription factor 1 Homo sapiens 51-56 32885522-4 2020 Sunitinib is the first oral indolin-2-one derivative marketed as a VEGFR inhibitor in the treatment of renal cell carcinoma and gastrointestinal stromal tumors. Sunitinib 0-9 kinase insert domain receptor Homo sapiens 67-72 33145941-2 2020 Here, we investigated whether the resistance to sunitinib (Sun), the standard treatment for metastatic ccRCC, is due to up-regulation of programmed death ligand 1 (PD-L1) by the transcription factor E3 (TFE3). Sunitinib 59-62 transcription factor binding to IGHM enhancer 3 Homo sapiens 178-201 33145941-2 2020 Here, we investigated whether the resistance to sunitinib (Sun), the standard treatment for metastatic ccRCC, is due to up-regulation of programmed death ligand 1 (PD-L1) by the transcription factor E3 (TFE3). Sunitinib 59-62 transcription factor binding to IGHM enhancer 3 Homo sapiens 203-207 33145941-0 2020 TFE3-PD-L1 axis is pivotal for sunitinib resistance in clear cell renal cell carcinoma. Sunitinib 31-40 CD274 molecule Homo sapiens 5-10 33145941-2 2020 Here, we investigated whether the resistance to sunitinib (Sun), the standard treatment for metastatic ccRCC, is due to up-regulation of programmed death ligand 1 (PD-L1) by the transcription factor E3 (TFE3). Sunitinib 48-57 CD274 molecule Homo sapiens 137-162 33145941-2 2020 Here, we investigated whether the resistance to sunitinib (Sun), the standard treatment for metastatic ccRCC, is due to up-regulation of programmed death ligand 1 (PD-L1) by the transcription factor E3 (TFE3). Sunitinib 48-57 CD274 molecule Homo sapiens 164-169 33145941-2 2020 Here, we investigated whether the resistance to sunitinib (Sun), the standard treatment for metastatic ccRCC, is due to up-regulation of programmed death ligand 1 (PD-L1) by the transcription factor E3 (TFE3). Sunitinib 48-57 transcription factor binding to IGHM enhancer 3 Homo sapiens 178-201 33145941-2 2020 Here, we investigated whether the resistance to sunitinib (Sun), the standard treatment for metastatic ccRCC, is due to up-regulation of programmed death ligand 1 (PD-L1) by the transcription factor E3 (TFE3). Sunitinib 48-57 transcription factor binding to IGHM enhancer 3 Homo sapiens 203-207 33145941-2 2020 Here, we investigated whether the resistance to sunitinib (Sun), the standard treatment for metastatic ccRCC, is due to up-regulation of programmed death ligand 1 (PD-L1) by the transcription factor E3 (TFE3). Sunitinib 59-62 CD274 molecule Homo sapiens 137-162 33145941-2 2020 Here, we investigated whether the resistance to sunitinib (Sun), the standard treatment for metastatic ccRCC, is due to up-regulation of programmed death ligand 1 (PD-L1) by the transcription factor E3 (TFE3). Sunitinib 59-62 CD274 molecule Homo sapiens 164-169 33261151-9 2020 In addition, we inoculated 786-O cells to generate xenografted mice to evaluate the anti-tumor activity of AJ-HP-beta-CD in vivo and found that AJ-HP-beta-CD had a better tumor inhibitory effect than that of docetaxel and sunitinib in terms of tumor growth and endpoint tumor weight. Sunitinib 222-231 adrenocortical dysplasia Mus musculus 150-157 33510997-0 2021 The Beneficial Role of Sunitinib in Tumor Immune Surveillance by Regulating Tumor PD-L1. Sunitinib 23-32 CD274 molecule Homo sapiens 82-87 33510997-4 2021 Mechanistically, it is discovered that Sunitinib regulates the stability of tumor PD-L1 via p62, that p62 can bind to PD-L1 and specifically promote its translocation into autophagic lysosome for degradation. Sunitinib 39-48 CD274 molecule Homo sapiens 82-87 33510997-4 2021 Mechanistically, it is discovered that Sunitinib regulates the stability of tumor PD-L1 via p62, that p62 can bind to PD-L1 and specifically promote its translocation into autophagic lysosome for degradation. Sunitinib 39-48 nucleoporin 62 Homo sapiens 92-95 33510997-4 2021 Mechanistically, it is discovered that Sunitinib regulates the stability of tumor PD-L1 via p62, that p62 can bind to PD-L1 and specifically promote its translocation into autophagic lysosome for degradation. Sunitinib 39-48 CD274 molecule Homo sapiens 118-123 33510997-5 2021 Preclinically, Sunitinib shows a synergistic antitumor effect with cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) monoclonal antibody (mAb) in melanoma and nonsmall cell lung cancer (NSCLC) immune competent mice by promoting the tumor-infiltrating lymphocytes activity. Sunitinib 15-24 cytotoxic T-lymphocyte-associated protein 4 Mus musculus 112-118 33510997-7 2021 Taken together, by utilizing rigorous computational analysis, functional characterization in vitro and in vivo, and neoadjuvent clinical trial, a novel molecular mechanism is revealed regarding the regulation of PD-L1 via p62, thus providing a novel therapeutic strategy by the combination treatment of CTLA-4 with Sunitinib. Sunitinib 315-324 CD274 molecule Homo sapiens 212-217 33510997-7 2021 Taken together, by utilizing rigorous computational analysis, functional characterization in vitro and in vivo, and neoadjuvent clinical trial, a novel molecular mechanism is revealed regarding the regulation of PD-L1 via p62, thus providing a novel therapeutic strategy by the combination treatment of CTLA-4 with Sunitinib. Sunitinib 315-324 nucleoporin 62 Homo sapiens 222-225 33510997-7 2021 Taken together, by utilizing rigorous computational analysis, functional characterization in vitro and in vivo, and neoadjuvent clinical trial, a novel molecular mechanism is revealed regarding the regulation of PD-L1 via p62, thus providing a novel therapeutic strategy by the combination treatment of CTLA-4 with Sunitinib. Sunitinib 315-324 cytotoxic T-lymphocyte associated protein 4 Homo sapiens 303-309 33068284-0 2020 Sunitinib in Patients with Metastatic Colorectal Cancer (mCRC) with FLT-3 Amplification: Results from the Targeted Agent and Profiling Utilization Registry (TAPUR) Study. Sunitinib 0-9 fms related receptor tyrosine kinase 3 Homo sapiens 68-73 33068284-2 2020 Sunitinib is an oral multikinase inhibitor of FMS-like tyrosine kinase-3 (FLT-3), among other targets. Sunitinib 0-9 fms related receptor tyrosine kinase 3 Homo sapiens 46-72 33068284-2 2020 Sunitinib is an oral multikinase inhibitor of FMS-like tyrosine kinase-3 (FLT-3), among other targets. Sunitinib 0-9 fms related receptor tyrosine kinase 3 Homo sapiens 74-79 33068284-3 2020 Results from a cohort of patients with metastatic colorectal cancer (mCRC) with FLT-3 amplification treated with sunitinib are reported. Sunitinib 113-122 fms related receptor tyrosine kinase 3 Homo sapiens 80-85 33068284-4 2020 OBJECTIVE: This study aimed to investigate whether patients with mCRC with FLT-3 amplification would be responsive to sunitinib, an oral multikinase inhibitor. Sunitinib 118-127 fms related receptor tyrosine kinase 3 Homo sapiens 75-80 33201837-0 2020 Low DAPK1 expression correlates with poor prognosis and sunitinib resistance in clear cell renal cell carcinoma. Sunitinib 56-65 death associated protein kinase 1 Homo sapiens 4-9 33218150-0 2020 Sunitinib-Containing Carborane Pharmacophore with the Ability to Inhibit Tyrosine Kinases Receptors FLT3, KIT and PDGFR-beta, Exhibits Powerful In Vivo Anti-Glioblastoma Activity. Sunitinib 0-9 fms related receptor tyrosine kinase 3 Homo sapiens 100-104 33218150-0 2020 Sunitinib-Containing Carborane Pharmacophore with the Ability to Inhibit Tyrosine Kinases Receptors FLT3, KIT and PDGFR-beta, Exhibits Powerful In Vivo Anti-Glioblastoma Activity. Sunitinib 0-9 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 106-109 33218150-0 2020 Sunitinib-Containing Carborane Pharmacophore with the Ability to Inhibit Tyrosine Kinases Receptors FLT3, KIT and PDGFR-beta, Exhibits Powerful In Vivo Anti-Glioblastoma Activity. Sunitinib 0-9 platelet derived growth factor receptor alpha Homo sapiens 114-124 33212804-0 2020 Small-Dose Sunitinib Modulates p53, Bcl-2, STAT3, and ERK1/2 Pathways and Protects against Adenine-Induced Nephrotoxicity. Sunitinib 11-20 transformation related protein 53, pseudogene Mus musculus 31-34 33212804-0 2020 Small-Dose Sunitinib Modulates p53, Bcl-2, STAT3, and ERK1/2 Pathways and Protects against Adenine-Induced Nephrotoxicity. Sunitinib 11-20 B cell leukemia/lymphoma 2 Mus musculus 36-41 33212804-0 2020 Small-Dose Sunitinib Modulates p53, Bcl-2, STAT3, and ERK1/2 Pathways and Protects against Adenine-Induced Nephrotoxicity. Sunitinib 11-20 signal transducer and activator of transcription 3 Mus musculus 43-48 33299657-0 2020 Soluble forms of PD-L1 and PD-1 as prognostic and predictive markers of sunitinib efficacy in patients with metastatic clear cell renal cell carcinoma. Sunitinib 72-81 CD274 molecule Homo sapiens 17-22 33299657-0 2020 Soluble forms of PD-L1 and PD-1 as prognostic and predictive markers of sunitinib efficacy in patients with metastatic clear cell renal cell carcinoma. Sunitinib 72-81 programmed cell death 1 Homo sapiens 27-31 33299657-8 2020 Patients with high T0 plasmatic levels of sPD-L1 had a shorter PFS (11.3 vs 22.5 months, p = .011) in the sunitinib group. Sunitinib 106-115 spindle apparatus coiled-coil protein 1 Homo sapiens 42-48 33299657-10 2020 mccRCC patients with high plasmatic levels of sPD-L1 or sPD-1 are poor responders to sunitinib. Sunitinib 85-94 spindle apparatus coiled-coil protein 1 Homo sapiens 46-52 33299657-10 2020 mccRCC patients with high plasmatic levels of sPD-L1 or sPD-1 are poor responders to sunitinib. Sunitinib 85-94 homeobox D13 Homo sapiens 56-61 33203399-9 2020 Grade 3/4 haematological toxicities were lower with docetaxel-sunitinib (18.2% vs 34.3%, p = 0.132), attributed to greater discretionary use of prophylactic G-CSF (90.9% vs 63.0%, p = 0.024). Sunitinib 62-71 colony stimulating factor 3 Homo sapiens 157-162 33212804-0 2020 Small-Dose Sunitinib Modulates p53, Bcl-2, STAT3, and ERK1/2 Pathways and Protects against Adenine-Induced Nephrotoxicity. Sunitinib 11-20 mitogen-activated protein kinase 3 Mus musculus 54-60 33201837-7 2020 Sunitinib-resistant ccRCC cells show significantly lower DAPK1 mRNA and protein levels than sunitinib-sensitive ccRCC cells. Sunitinib 0-9 death associated protein kinase 1 Homo sapiens 57-62 33212804-3 2020 To test our hypothesis, we investigated sunitinib as an inhibitor for tyrosine kinase signaling for both vascular endothelial growth factor receptor (VEGFR) and platelet-derived growth factor receptors (PDGFR) against adenine-induced nephrotoxicity. Sunitinib 40-49 platelet derived growth factor receptor, beta polypeptide Mus musculus 161-201 33212804-3 2020 To test our hypothesis, we investigated sunitinib as an inhibitor for tyrosine kinase signaling for both vascular endothelial growth factor receptor (VEGFR) and platelet-derived growth factor receptors (PDGFR) against adenine-induced nephrotoxicity. Sunitinib 40-49 platelet derived growth factor receptor, beta polypeptide Mus musculus 203-208 33201837-8 2020 DAPK1 overexpression enhances apoptosis in sunitinib-resistant ccRCC cells via the ATF6-dependent ER stress pathway. Sunitinib 43-52 death associated protein kinase 1 Homo sapiens 0-5 33201837-8 2020 DAPK1 overexpression enhances apoptosis in sunitinib-resistant ccRCC cells via the ATF6-dependent ER stress pathway. Sunitinib 43-52 activating transcription factor 6 Homo sapiens 83-87 33212804-10 2020 Furthermore, sunitinib decreased (p < 0.5) renal levels of TGF-beta1, p-ERK1/2, and phospho-STAT3 while elevating Bcl-2 expression score. Sunitinib 13-22 transforming growth factor, beta 1 Mus musculus 59-68 33201837-10 2020 Our finding demonstrates that low DAPK1 expression is an independent prognostic indicator that correlates with ccRCC progression and sunitinib resistance. Sunitinib 133-142 death associated protein kinase 1 Mus musculus 34-39 33212804-10 2020 Furthermore, sunitinib decreased (p < 0.5) renal levels of TGF-beta1, p-ERK1/2, and phospho-STAT3 while elevating Bcl-2 expression score. Sunitinib 13-22 mitogen-activated protein kinase 3 Mus musculus 72-78 33212804-10 2020 Furthermore, sunitinib decreased (p < 0.5) renal levels of TGF-beta1, p-ERK1/2, and phospho-STAT3 while elevating Bcl-2 expression score. Sunitinib 13-22 signal transducer and activator of transcription 3 Mus musculus 92-97 33212804-10 2020 Furthermore, sunitinib decreased (p < 0.5) renal levels of TGF-beta1, p-ERK1/2, and phospho-STAT3 while elevating Bcl-2 expression score. Sunitinib 13-22 B cell leukemia/lymphoma 2 Mus musculus 114-119 33083006-0 2020 Sunitinib reduces the infection of SARS-CoV, MERS-CoV and SARS-CoV-2 partially by inhibiting AP2M1 phosphorylation. Sunitinib 0-9 adaptor related protein complex 2 subunit mu 1 Homo sapiens 93-98 32901843-4 2020 In the present study, we examined which splice variants of LAMP-2 contributed to sunitinib resistance in RCC cells. Sunitinib 81-90 lysosomal associated membrane protein 2 Homo sapiens 59-65 32761670-0 2020 Sunitinib induces primary ectopic endometrial cell apoptosis through up-regulation of STAT1 in vitro. Sunitinib 0-9 signal transducer and activator of transcription 1 Homo sapiens 86-91 32761670-11 2020 Protein spectrum analysis was conducted on ectopic intimal cells after sunitinib treatment, and it was found that STAT1 is specifically expressed in ectopic endometrial cells. Sunitinib 71-80 signal transducer and activator of transcription 1 Homo sapiens 114-119 32761670-12 2020 In vitro, and through fludarabine interference, it was revealed that sunitinib specifically inhibited the phosphorylation site Tyr751 of PDGFR, while the expression of STAT1, p-STAT1, and caspase-3 was significantly upregulated, and the expression of STAT1 and p-STAT1 was positively correlated with the expression of caspase-3. Sunitinib 69-78 platelet derived growth factor receptor beta Homo sapiens 137-142 32761670-12 2020 In vitro, and through fludarabine interference, it was revealed that sunitinib specifically inhibited the phosphorylation site Tyr751 of PDGFR, while the expression of STAT1, p-STAT1, and caspase-3 was significantly upregulated, and the expression of STAT1 and p-STAT1 was positively correlated with the expression of caspase-3. Sunitinib 69-78 signal transducer and activator of transcription 1 Homo sapiens 168-173 32761670-12 2020 In vitro, and through fludarabine interference, it was revealed that sunitinib specifically inhibited the phosphorylation site Tyr751 of PDGFR, while the expression of STAT1, p-STAT1, and caspase-3 was significantly upregulated, and the expression of STAT1 and p-STAT1 was positively correlated with the expression of caspase-3. Sunitinib 69-78 signal transducer and activator of transcription 1 Homo sapiens 177-182 32761670-12 2020 In vitro, and through fludarabine interference, it was revealed that sunitinib specifically inhibited the phosphorylation site Tyr751 of PDGFR, while the expression of STAT1, p-STAT1, and caspase-3 was significantly upregulated, and the expression of STAT1 and p-STAT1 was positively correlated with the expression of caspase-3. Sunitinib 69-78 caspase 3 Homo sapiens 188-197 32761670-12 2020 In vitro, and through fludarabine interference, it was revealed that sunitinib specifically inhibited the phosphorylation site Tyr751 of PDGFR, while the expression of STAT1, p-STAT1, and caspase-3 was significantly upregulated, and the expression of STAT1 and p-STAT1 was positively correlated with the expression of caspase-3. Sunitinib 69-78 signal transducer and activator of transcription 1 Homo sapiens 177-182 32761670-12 2020 In vitro, and through fludarabine interference, it was revealed that sunitinib specifically inhibited the phosphorylation site Tyr751 of PDGFR, while the expression of STAT1, p-STAT1, and caspase-3 was significantly upregulated, and the expression of STAT1 and p-STAT1 was positively correlated with the expression of caspase-3. Sunitinib 69-78 signal transducer and activator of transcription 1 Homo sapiens 177-182 32761670-12 2020 In vitro, and through fludarabine interference, it was revealed that sunitinib specifically inhibited the phosphorylation site Tyr751 of PDGFR, while the expression of STAT1, p-STAT1, and caspase-3 was significantly upregulated, and the expression of STAT1 and p-STAT1 was positively correlated with the expression of caspase-3. Sunitinib 69-78 caspase 3 Homo sapiens 318-327 32761670-14 2020 CONCLUSION: The in vitro experiments revealed that sunitinib could upregulate the expression of STAT1 by inhibiting the phosphorylation site Tyr751 of PDGFR, thereby specifically inducing the apoptosis of the primary heterotopic mesenchymal endometrium. Sunitinib 51-60 signal transducer and activator of transcription 1 Homo sapiens 96-101 32761670-14 2020 CONCLUSION: The in vitro experiments revealed that sunitinib could upregulate the expression of STAT1 by inhibiting the phosphorylation site Tyr751 of PDGFR, thereby specifically inducing the apoptosis of the primary heterotopic mesenchymal endometrium. Sunitinib 51-60 platelet derived growth factor receptor beta Homo sapiens 151-156 32879446-1 2020 Angiogenesis inhibitors, such as the receptor tyrosine kinase (RTK) inhibitor sunitinib, target vascular endothelial growth factor (VEGF) signaling in cancers. Sunitinib 78-87 TYRO3 protein tyrosine kinase 3 Mus musculus 37-61 33041663-9 2020 In vitro validation showed RBL1 overexpression induced resistance to Sunitinib and Cabozantinib; KLHL33 silencing induced resistance to Sunitinib; ARHGAP28 silencing induced sensitivity to Cabozantinib. Sunitinib 69-78 RB transcriptional corepressor like 1 Homo sapiens 27-31 33041663-9 2020 In vitro validation showed RBL1 overexpression induced resistance to Sunitinib and Cabozantinib; KLHL33 silencing induced resistance to Sunitinib; ARHGAP28 silencing induced sensitivity to Cabozantinib. Sunitinib 136-145 kelch like family member 33 Homo sapiens 97-103 32788078-6 2020 The observed sunitinib concentrations were a little higher than those reported in the 421C>A polymorphism of the ATP-binding cassette subfamily G member 2 gene carrier. Sunitinib 13-22 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 113-154 33115942-8 2020 We found that RFA caused PD-1 upregulation in tumor-infiltrated T cells by boosting hepatocyte growth factor (HGF) expression, which was suppressed by sunitinib treatment. Sunitinib 151-160 hepatocyte growth factor Mus musculus 84-108 33115942-8 2020 We found that RFA caused PD-1 upregulation in tumor-infiltrated T cells by boosting hepatocyte growth factor (HGF) expression, which was suppressed by sunitinib treatment. Sunitinib 151-160 hepatocyte growth factor Mus musculus 110-113 33115942-9 2020 We have also demonstrated that sunitinib suppressed VEGF"s effect in enhancing PD-L1 expression in DCs and attenuated heat-sink effect. Sunitinib 31-40 vascular endothelial growth factor A Mus musculus 52-56 33115942-9 2020 We have also demonstrated that sunitinib suppressed VEGF"s effect in enhancing PD-L1 expression in DCs and attenuated heat-sink effect. Sunitinib 31-40 CD274 antigen Mus musculus 79-84 33115942-11 2020 CONCLUSIONS: Sunitinib enables RFA-released in situ TSA to ignite an effective anti-tumor immune response by suppressing HGF and VEGF signaling pathways. Sunitinib 13-22 hepatocyte growth factor Mus musculus 121-124 33115942-11 2020 CONCLUSIONS: Sunitinib enables RFA-released in situ TSA to ignite an effective anti-tumor immune response by suppressing HGF and VEGF signaling pathways. Sunitinib 13-22 vascular endothelial growth factor A Mus musculus 129-133 32229739-12 2020 RESULTS: Under sunitinib therapy, assessment of the expression of VEGFR2 by ultrasound molecular imaging with BR55 reveals a significant difference as early as 12 hours after first dosing (-25%), whereas tumor size significant change occurs only after 24 hours. Sunitinib 15-24 kinase insert domain receptor Rattus norvegicus 66-72 32879446-1 2020 Angiogenesis inhibitors, such as the receptor tyrosine kinase (RTK) inhibitor sunitinib, target vascular endothelial growth factor (VEGF) signaling in cancers. Sunitinib 78-87 TYRO3 protein tyrosine kinase 3 Mus musculus 63-66 32879446-1 2020 Angiogenesis inhibitors, such as the receptor tyrosine kinase (RTK) inhibitor sunitinib, target vascular endothelial growth factor (VEGF) signaling in cancers. Sunitinib 78-87 vascular endothelial growth factor A Mus musculus 96-130 32879446-1 2020 Angiogenesis inhibitors, such as the receptor tyrosine kinase (RTK) inhibitor sunitinib, target vascular endothelial growth factor (VEGF) signaling in cancers. Sunitinib 78-87 vascular endothelial growth factor A Mus musculus 132-136 32879446-7 2020 EC-specific PI3Kbeta knockout (EpsilonC-betaKO) in mice potentiated the sunitinib-induced reduction in subcutaneous growth of LLC1 and B16F10, and lung metastasis of B16F10 tumors. Sunitinib 72-81 phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit beta Mus musculus 12-20 32879446-10 2020 Taken together, EC PI3Kbeta inactivation with sunitinib inhibition reduces microvessel turnover and decreases heterogeneity of the tumor microenvironment, hence PI3Kbeta inhibition may be a useful adjuvant antiangiogenesis therapy with sunitinib. Sunitinib 46-55 phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit beta Mus musculus 19-27 32879446-10 2020 Taken together, EC PI3Kbeta inactivation with sunitinib inhibition reduces microvessel turnover and decreases heterogeneity of the tumor microenvironment, hence PI3Kbeta inhibition may be a useful adjuvant antiangiogenesis therapy with sunitinib. Sunitinib 236-245 phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit beta Mus musculus 19-27 32546645-1 2020 PURPOSE: In the S-TRAC trial, adjuvant sunitinib improved disease-free survival (DFS) compared with placebo in patients with loco-regional renal cell carcinoma (RCC) at high risk of recurrence. Sunitinib 39-48 T cell receptor alpha constant Homo sapiens 18-22 32248228-3 2020 The objective of this study was to examine whether SAL enhances the cytotoxic effect of sunitinib in Gb3-expressing HeLa cells. Sunitinib 88-97 alpha 1,4-galactosyltransferase (P blood group) Homo sapiens 101-104 32248228-8 2020 Furthermore, we observed that SAL promoted the formation of lysosome-like structures, which are LAMP1 positive but not acidic in HeLa cells, that can trap sunitinib. Sunitinib 155-164 lysosomal associated membrane protein 1 Homo sapiens 96-101 32248228-10 2020 Our findings suggest that SAL/Gb3 interaction promoted sunitinib uptake and suppressed sunitinib excretion, and that sunitinib efficiently exerted cytotoxicity against HeLa cells. Sunitinib 55-64 alpha 1,4-galactosyltransferase (P blood group) Homo sapiens 30-33 32248228-10 2020 Our findings suggest that SAL/Gb3 interaction promoted sunitinib uptake and suppressed sunitinib excretion, and that sunitinib efficiently exerted cytotoxicity against HeLa cells. Sunitinib 87-96 alpha 1,4-galactosyltransferase (P blood group) Homo sapiens 30-33 32248228-10 2020 Our findings suggest that SAL/Gb3 interaction promoted sunitinib uptake and suppressed sunitinib excretion, and that sunitinib efficiently exerted cytotoxicity against HeLa cells. Sunitinib 87-96 alpha 1,4-galactosyltransferase (P blood group) Homo sapiens 30-33 33072561-0 2020 Long Non-coding RNA CCAT1 Acts as an Oncogene and Promotes Sunitinib Resistance in Renal Cell Carcinoma. Sunitinib 59-68 colon cancer associated transcript 1 Homo sapiens 20-25 33072561-3 2020 We found long non-coding RNA (lncRNA) colon cancer-associated transcript-1 (CCAT1) overexpressed in sunitinib-resistant cells while declined in the parental cells. Sunitinib 100-109 colon cancer associated transcript 1 Homo sapiens 76-81 33072561-4 2020 Moreover, lncRNA CCAT1 increased significantly in samples with resistance to sunitinib compared with those with responses to sunitinib. Sunitinib 77-86 colon cancer associated transcript 1 Homo sapiens 17-22 33072561-8 2020 Apart from the in vitro experiments, we demonstrated that knockdown of CCAT1 boosted response to sunitinib by performing sunitinib-resistant ACHN mouse models. Sunitinib 97-106 colon cancer associated transcript 1 Homo sapiens 71-76 33072561-8 2020 Apart from the in vitro experiments, we demonstrated that knockdown of CCAT1 boosted response to sunitinib by performing sunitinib-resistant ACHN mouse models. Sunitinib 121-130 colon cancer associated transcript 1 Homo sapiens 71-76 33072561-9 2020 Briefly, lncRNA CCAT1 conferred renal cell carcinoma resistance to sunitinib in a c-Myc-dependent manner, providing a novel target for improvement of sunitinib therapy. Sunitinib 67-76 colon cancer associated transcript 1 Homo sapiens 16-21 33072561-9 2020 Briefly, lncRNA CCAT1 conferred renal cell carcinoma resistance to sunitinib in a c-Myc-dependent manner, providing a novel target for improvement of sunitinib therapy. Sunitinib 67-76 MYC proto-oncogene, bHLH transcription factor Homo sapiens 82-87 33072561-9 2020 Briefly, lncRNA CCAT1 conferred renal cell carcinoma resistance to sunitinib in a c-Myc-dependent manner, providing a novel target for improvement of sunitinib therapy. Sunitinib 150-159 colon cancer associated transcript 1 Homo sapiens 16-21 33072561-9 2020 Briefly, lncRNA CCAT1 conferred renal cell carcinoma resistance to sunitinib in a c-Myc-dependent manner, providing a novel target for improvement of sunitinib therapy. Sunitinib 150-159 MYC proto-oncogene, bHLH transcription factor Homo sapiens 82-87 32830849-0 2020 Sunitinib inhibits RNase L by destabilizing its active dimer conformation. Sunitinib 0-9 ribonuclease L Homo sapiens 19-26 32830849-2 2020 The ribonuclease activity of RNase L can be regulated by the kinase inhibitor sunitinib. Sunitinib 78-87 ribonuclease L Homo sapiens 29-36 32830849-4 2020 In this study, we aimed to uncover the mechanism of action through which sunitinib inhibits RNase L. Sunitinib 73-82 ribonuclease L Homo sapiens 92-99 32830849-5 2020 We solved the crystal structures of RNase L in complex with sunitinib and its analogs toceranib and SU11652. Sunitinib 60-69 ribonuclease L Homo sapiens 36-43 32830849-6 2020 Our results showed that sunitinib bound to the ATP-binding pocket of RNase L. Sunitinib 24-33 ribonuclease L Homo sapiens 69-76 32830849-8 2020 Molecular dynamics simulations and dynamic light scattering results support that the binding of sunitinib in the pseudokinase domain destabilized the dimer conformation of RNase L and allosterically inhibited its ribonuclease activity. Sunitinib 96-105 ribonuclease L Homo sapiens 172-179 32993785-1 2020 BACKGROUND: Sunitinib, a receptor tyrosine kinase (RTK) inhibitor that targets multiple receptors such as vascular endothelial growth factor receptors (VEGFRs), was approved for cancer treatment in 2006. Sunitinib 12-21 TYRO3 protein tyrosine kinase 3 Mus musculus 25-49 32993785-1 2020 BACKGROUND: Sunitinib, a receptor tyrosine kinase (RTK) inhibitor that targets multiple receptors such as vascular endothelial growth factor receptors (VEGFRs), was approved for cancer treatment in 2006. Sunitinib 12-21 TYRO3 protein tyrosine kinase 3 Mus musculus 51-54 32921632-0 2020 LINC00160 mediates sunitinib resistance in renal cell carcinoma via SAA1 that is implicated in STAT3 activation and compound transportation. Sunitinib 19-28 long intergenic non-protein coding RNA 160 Homo sapiens 0-9 33052225-0 2020 Restoring the epigenetically silenced PCK2 suppresses renal cell carcinoma progression and increases sensitivity to sunitinib by promoting endoplasmic reticulum stress. Sunitinib 116-125 phosphoenolpyruvate carboxykinase 2, mitochondrial Homo sapiens 38-42 33052225-11 2020 Restoration of PCK2 expression in RCC cells repressed tumor progression and increased their sensitivity to sunitinib. Sunitinib 107-116 phosphoenolpyruvate carboxykinase 2, mitochondrial Homo sapiens 15-19 33052225-13 2020 Conclusions: Collectively, our results identify a specific mechanism by which PCK2 suppresses the progression of renal cell carcinoma (RCC) and increases sensitivity to sunitinib by promoting endoplasmic reticulum stress. Sunitinib 169-178 phosphoenolpyruvate carboxykinase 2, mitochondrial Homo sapiens 78-82 32921632-0 2020 LINC00160 mediates sunitinib resistance in renal cell carcinoma via SAA1 that is implicated in STAT3 activation and compound transportation. Sunitinib 19-28 serum amyloid A1 Homo sapiens 68-72 32921632-3 2020 In this study, we identified that LINC00160 was associated with sunitinib resistance in renal cell carcinoma. Sunitinib 64-73 long intergenic non-protein coding RNA 160 Homo sapiens 34-43 32921632-5 2020 On one hand, SAA1 linked to ABCB1 protein, which facilitated sunitinib cellular efflux and diminished drug accumulation. Sunitinib 61-70 serum amyloid A1 Homo sapiens 13-17 32921632-5 2020 On one hand, SAA1 linked to ABCB1 protein, which facilitated sunitinib cellular efflux and diminished drug accumulation. Sunitinib 61-70 ATP binding cassette subfamily B member 1 Homo sapiens 28-33 32921632-8 2020 LINC00160 mediates sunitinib resistance in renal cell carcinoma via SAA1 that is implicated in STAT3 activation and compound transportation, which offers an opportunity for targeted intervention and molecular therapies in the future. Sunitinib 19-28 long intergenic non-protein coding RNA 160 Homo sapiens 0-9 32921632-8 2020 LINC00160 mediates sunitinib resistance in renal cell carcinoma via SAA1 that is implicated in STAT3 activation and compound transportation, which offers an opportunity for targeted intervention and molecular therapies in the future. Sunitinib 19-28 serum amyloid A1 Homo sapiens 68-72 32552305-0 2020 Third-line sunitinib treatment in a VHL-mutated metastatic intrahepatic cholangiocarcinoma: a case report and literature review. Sunitinib 11-20 von Hippel-Lindau tumor suppressor Homo sapiens 36-39 32552305-5 2020 According to next-generation sequencing result of somatic Von Hippel-Lindau (VHL) gene mutation, the patient was administered third-line sunitinib and obtained a relatively longer survival of 9 months after taking sunitinib. Sunitinib 137-146 von Hippel-Lindau tumor suppressor Homo sapiens 58-75 32552305-5 2020 According to next-generation sequencing result of somatic Von Hippel-Lindau (VHL) gene mutation, the patient was administered third-line sunitinib and obtained a relatively longer survival of 9 months after taking sunitinib. Sunitinib 137-146 von Hippel-Lindau tumor suppressor Homo sapiens 77-80 32552305-5 2020 According to next-generation sequencing result of somatic Von Hippel-Lindau (VHL) gene mutation, the patient was administered third-line sunitinib and obtained a relatively longer survival of 9 months after taking sunitinib. Sunitinib 214-223 von Hippel-Lindau tumor suppressor Homo sapiens 58-75 32947305-4 2020 Evidence supporting the use of these therapies in the adjuvant setting is mixed, although one clinical trial, S-TRAC, has shown improvements in disease-free survival with 1 year of adjuvant sunitinib among patients with clear cell histology and high-risk features, leading to the first US Food and Drug Administration approval of an adjuvant therapy for high-risk RCC patients. Sunitinib 190-199 T cell receptor alpha constant Homo sapiens 112-116 32552305-5 2020 According to next-generation sequencing result of somatic Von Hippel-Lindau (VHL) gene mutation, the patient was administered third-line sunitinib and obtained a relatively longer survival of 9 months after taking sunitinib. Sunitinib 214-223 von Hippel-Lindau tumor suppressor Homo sapiens 77-80 32814829-0 2020 Enhanced YB1/EphA2 axis signaling promotes acquired resistance to sunitinib and metastatic potential in renal cell carcinoma. Sunitinib 66-75 Y-box binding protein 1 Homo sapiens 9-12 32291709-2 2020 Imatinib and sunitinib are approved KIT-inhibiting therapies. Sunitinib 13-22 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 36-39 32291709-9 2020 Our data show that dasatinib is more potent than imatinib or sunitinib at inhibiting the activity of drug-resistant KIT mutants. Sunitinib 61-70 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 116-119 32783497-0 2020 Feedback activation of GATA1/miR-885-5p/PLIN3 pathway decreases sunitinib sensitivity in clear cell renal cell carcinoma. Sunitinib 64-73 GATA binding protein 1 Homo sapiens 23-28 32783497-0 2020 Feedback activation of GATA1/miR-885-5p/PLIN3 pathway decreases sunitinib sensitivity in clear cell renal cell carcinoma. Sunitinib 64-73 microRNA 885 Homo sapiens 29-36 32783497-0 2020 Feedback activation of GATA1/miR-885-5p/PLIN3 pathway decreases sunitinib sensitivity in clear cell renal cell carcinoma. Sunitinib 64-73 perilipin 3 Homo sapiens 40-45 32783497-3 2020 Here, we discovered miR-885-5p was notably decreased after sunitinib treatment and associated with poor disease progression in clear cell renal cell carcinoma (ccRCC). Sunitinib 59-68 microRNA 885 Homo sapiens 20-27 32783497-5 2020 Mechanistically, sunitinib treatment reduced GATA1 expression, which in turn reduced its binding to MIR885 promoter and resulted in miR-885-5p downregulation in transcriptional level. Sunitinib 17-26 GATA binding protein 1 Homo sapiens 45-50 32783497-5 2020 Mechanistically, sunitinib treatment reduced GATA1 expression, which in turn reduced its binding to MIR885 promoter and resulted in miR-885-5p downregulation in transcriptional level. Sunitinib 17-26 microRNA 885 Homo sapiens 100-106 32783497-5 2020 Mechanistically, sunitinib treatment reduced GATA1 expression, which in turn reduced its binding to MIR885 promoter and resulted in miR-885-5p downregulation in transcriptional level. Sunitinib 17-26 microRNA 885 Homo sapiens 132-139 32783497-6 2020 In addition, PLIN3 was confirmed to be directly targeted by miR-885-5p and its upregulation significantly increased lipid droplets formation to decrease sunitinib sensitivity. Sunitinib 153-162 perilipin 3 Homo sapiens 13-18 32783497-6 2020 In addition, PLIN3 was confirmed to be directly targeted by miR-885-5p and its upregulation significantly increased lipid droplets formation to decrease sunitinib sensitivity. Sunitinib 153-162 microRNA 885 Homo sapiens 60-67 32783497-7 2020 Therefore, GATA1/miR-885-5p/ PLIN3 pathway may serve as a potential therapeutic strategy and a biomarker for sunitinib treatment in ccRCC. Sunitinib 109-118 GATA binding protein 1 Homo sapiens 11-16 32783497-7 2020 Therefore, GATA1/miR-885-5p/ PLIN3 pathway may serve as a potential therapeutic strategy and a biomarker for sunitinib treatment in ccRCC. Sunitinib 109-118 microRNA 885 Homo sapiens 17-24 32783497-7 2020 Therefore, GATA1/miR-885-5p/ PLIN3 pathway may serve as a potential therapeutic strategy and a biomarker for sunitinib treatment in ccRCC. Sunitinib 109-118 perilipin 3 Homo sapiens 29-34 32814829-5 2020 In addition, our findings confirm that YB1 promotes the invasion, metastasis and sunitinib resistance of ccRCC by regulating the EphA2 signaling pathway. Sunitinib 81-90 Y-box binding protein 1 Homo sapiens 39-42 32814829-6 2020 Furthermore, pharmacological inhibition of EphA2 through the small molecule inhibitor ALW-II-41-27 reduced the proliferation of sunitinib-resistant tumor cells, suppressed tumor growth in vivo, and restored the sensitivity of sunitinib-resistant tumor cells to sunitinib in vitro and in vivo. Sunitinib 128-137 EPH receptor A2 Homo sapiens 43-48 32814829-6 2020 Furthermore, pharmacological inhibition of EphA2 through the small molecule inhibitor ALW-II-41-27 reduced the proliferation of sunitinib-resistant tumor cells, suppressed tumor growth in vivo, and restored the sensitivity of sunitinib-resistant tumor cells to sunitinib in vitro and in vivo. Sunitinib 226-235 EPH receptor A2 Homo sapiens 43-48 32814829-6 2020 Furthermore, pharmacological inhibition of EphA2 through the small molecule inhibitor ALW-II-41-27 reduced the proliferation of sunitinib-resistant tumor cells, suppressed tumor growth in vivo, and restored the sensitivity of sunitinib-resistant tumor cells to sunitinib in vitro and in vivo. Sunitinib 226-235 EPH receptor A2 Homo sapiens 43-48 32814829-0 2020 Enhanced YB1/EphA2 axis signaling promotes acquired resistance to sunitinib and metastatic potential in renal cell carcinoma. Sunitinib 66-75 EPH receptor A2 Homo sapiens 13-18 32814829-2 2020 Receptor tyrosine kinase (RTK) inhibitors such as sunitinib have been demonstrated to target tumorigenic signaling pathways, delay tumor progression, and improve patient prognosis in metastatic renal cell carcinoma (mRCC). Sunitinib 50-59 ret proto-oncogene Homo sapiens 0-24 32814829-2 2020 Receptor tyrosine kinase (RTK) inhibitors such as sunitinib have been demonstrated to target tumorigenic signaling pathways, delay tumor progression, and improve patient prognosis in metastatic renal cell carcinoma (mRCC). Sunitinib 50-59 ret proto-oncogene Homo sapiens 26-29 32814829-4 2020 In our study, we found that increased expression of Y-box binding protein 1 (YB1, a multidrug resistance associated protein) and EphA2 (a member of the erythropoietin-producing hepatocellular (Eph) receptor family, belonging to the RTK family) mediated sunitinib resistance and mRCC exhibited a large phenotypic dependence on YB1 and EphA2. Sunitinib 253-262 Y-box binding protein 1 Homo sapiens 52-75 32814829-4 2020 In our study, we found that increased expression of Y-box binding protein 1 (YB1, a multidrug resistance associated protein) and EphA2 (a member of the erythropoietin-producing hepatocellular (Eph) receptor family, belonging to the RTK family) mediated sunitinib resistance and mRCC exhibited a large phenotypic dependence on YB1 and EphA2. Sunitinib 253-262 Y-box binding protein 1 Homo sapiens 77-80 32814829-4 2020 In our study, we found that increased expression of Y-box binding protein 1 (YB1, a multidrug resistance associated protein) and EphA2 (a member of the erythropoietin-producing hepatocellular (Eph) receptor family, belonging to the RTK family) mediated sunitinib resistance and mRCC exhibited a large phenotypic dependence on YB1 and EphA2. Sunitinib 253-262 ATP binding cassette subfamily C member 3 Homo sapiens 84-123 32814829-4 2020 In our study, we found that increased expression of Y-box binding protein 1 (YB1, a multidrug resistance associated protein) and EphA2 (a member of the erythropoietin-producing hepatocellular (Eph) receptor family, belonging to the RTK family) mediated sunitinib resistance and mRCC exhibited a large phenotypic dependence on YB1 and EphA2. Sunitinib 253-262 EPH receptor A2 Homo sapiens 129-134 32767163-4 2020 As the tumor may still be anti-VEGFR drug-naive, a tyrosine kinase inhibitor approved for first line treatment (e.g., sunitinib or pazopanib) may be beneficial. Sunitinib 118-127 kinase insert domain receptor Homo sapiens 31-36 32872567-7 2020 Furthermore, inhibition of AAK1 kinase activity using sunitinib decreased RABV infection. Sunitinib 54-63 AP2 associated kinase 1 Homo sapiens 27-31 32492594-5 2020 Six small molecules that inhibit VEGFR1/2/3, namely, sunitinib, sorafenib, axitinib, pazopanib, cabozantinib, and lenvatinib, have been approved by the Food and Drug Administration (FDA) for the treatment of RCC. Sunitinib 53-62 fms related receptor tyrosine kinase 1 Homo sapiens 33-43 32810161-0 2020 Cyclic pentapeptide cRGDfK enhances the inhibitory effect of sunitinib on TGF-beta1-induced epithelial-to-mesenchymal transition in human non-small cell lung cancer cells. Sunitinib 61-70 transforming growth factor beta 1 Homo sapiens 74-83 32810161-5 2020 Sunitinib strongly inhibited the TGF-beta1-activated EMT through suppression of Wnt signaling, Smad and non-Smad signaling pathways. Sunitinib 0-9 transforming growth factor beta 1 Homo sapiens 33-42 32874210-8 2020 Nivolumab plus ipilimumab and pembrolizumab plus axitinib achieved more health benefits than the other ICI regimens and sunitinib in programmed death ligand 1 (PD-L1)-positive and negative tumors, respectively. Sunitinib 120-129 CD274 molecule Homo sapiens 133-158 32874210-8 2020 Nivolumab plus ipilimumab and pembrolizumab plus axitinib achieved more health benefits than the other ICI regimens and sunitinib in programmed death ligand 1 (PD-L1)-positive and negative tumors, respectively. Sunitinib 120-129 CD274 molecule Homo sapiens 160-165 32628820-7 2020 CCNE1 gain was associated with resistance to Axitinib and LRP10 loss was associated with resistance to Sunitinib. Sunitinib 103-112 LDL receptor related protein 10 Homo sapiens 58-63 32923192-1 2020 Spontaneous pneumothorax secondary to sunitinib, a vascular endothelial growth factor receptor (VEGFR) inhibitor, is an extremely rare side effect of this class of medications. Sunitinib 38-47 kinase insert domain receptor Homo sapiens 51-94 32923192-1 2020 Spontaneous pneumothorax secondary to sunitinib, a vascular endothelial growth factor receptor (VEGFR) inhibitor, is an extremely rare side effect of this class of medications. Sunitinib 38-47 kinase insert domain receptor Homo sapiens 96-101 32578660-3 2020 Herein, the multi-targeted receptor tyrosine kinase (RTK) inhibitor sunitinib (Sun) and photodynamic therapy (PDT) drug chlorin e6 (Ce6) were locally delivered to the postoperative tumor site via a zwitterionic hydrogel. Sunitinib 68-77 ret proto-oncogene Homo sapiens 27-51 32578660-3 2020 Herein, the multi-targeted receptor tyrosine kinase (RTK) inhibitor sunitinib (Sun) and photodynamic therapy (PDT) drug chlorin e6 (Ce6) were locally delivered to the postoperative tumor site via a zwitterionic hydrogel. Sunitinib 68-77 ret proto-oncogene Homo sapiens 53-56 32663095-0 2020 Depletion of lncRNA MALAT1 inhibited sunitinib resistance through regulating miR-362-3p-mediated G3BP1 in renal cell carcinoma. Sunitinib 37-46 metastasis associated lung adenocarcinoma transcript 1 Homo sapiens 20-26 32663095-0 2020 Depletion of lncRNA MALAT1 inhibited sunitinib resistance through regulating miR-362-3p-mediated G3BP1 in renal cell carcinoma. Sunitinib 37-46 G3BP stress granule assembly factor 1 Homo sapiens 97-102 32663095-4 2020 Further, RNA pull-down and luciferase reporter assay were applied to investigate the underlying mechanism of MALAT1 in SU resistance. Sunitinib 119-121 metastasis associated lung adenocarcinoma transcript 1 Homo sapiens 109-115 32663095-2 2020 In this study, MALAT1 expression in SU-resistance tumor tissues and cells was tested by qRT-PCR. Sunitinib 36-38 metastasis associated lung adenocarcinoma transcript 1 Homo sapiens 15-21 32663095-5 2020 The results showed that MALAT1 expression was dramatically upregulated in SU-resistance RCC tissues and cell lines. Sunitinib 74-76 metastasis associated lung adenocarcinoma transcript 1 Homo sapiens 24-30 32760216-0 2020 LncRNA HOTAIR induces sunitinib resistance in renal cancer by acting as a competing endogenous RNA to regulate autophagy of renal cells. Sunitinib 22-31 HOX transcript antisense RNA (non-protein coding) Mus musculus 7-13 32663095-9 2020 Rescue experiments suggested that downregulation of miR-362-3p and overexpression of G3BP1 can reverse the SU chemosensitivity of MALAT1 knockdown in RCC cells. Sunitinib 107-109 G3BP stress granule assembly factor 1 Homo sapiens 85-90 32663095-9 2020 Rescue experiments suggested that downregulation of miR-362-3p and overexpression of G3BP1 can reverse the SU chemosensitivity of MALAT1 knockdown in RCC cells. Sunitinib 107-109 metastasis associated lung adenocarcinoma transcript 1 Homo sapiens 130-136 32663095-10 2020 In conclusion, depletion of LncRNA MALAT1 inhibited SU chemoresistance through modulating G3BP1 via sponging miR-362-3p in RCC cells, suggesting that targeting MALAT1 may be a potential therapeutic strategy for SU-resistance RCC. Sunitinib 52-54 metastasis associated lung adenocarcinoma transcript 1 Homo sapiens 35-41 32663095-10 2020 In conclusion, depletion of LncRNA MALAT1 inhibited SU chemoresistance through modulating G3BP1 via sponging miR-362-3p in RCC cells, suggesting that targeting MALAT1 may be a potential therapeutic strategy for SU-resistance RCC. Sunitinib 52-54 G3BP stress granule assembly factor 1 Homo sapiens 90-95 32663095-10 2020 In conclusion, depletion of LncRNA MALAT1 inhibited SU chemoresistance through modulating G3BP1 via sponging miR-362-3p in RCC cells, suggesting that targeting MALAT1 may be a potential therapeutic strategy for SU-resistance RCC. Sunitinib 211-213 metastasis associated lung adenocarcinoma transcript 1 Homo sapiens 35-41 32663095-10 2020 In conclusion, depletion of LncRNA MALAT1 inhibited SU chemoresistance through modulating G3BP1 via sponging miR-362-3p in RCC cells, suggesting that targeting MALAT1 may be a potential therapeutic strategy for SU-resistance RCC. Sunitinib 211-213 G3BP stress granule assembly factor 1 Homo sapiens 90-95 32511016-0 2020 Sunitinib inhibits PD-L1 expression in osteosarcoma by targeting STAT3 and remodels the immune system in tumor-bearing mice. Sunitinib 0-9 CD274 antigen Mus musculus 19-24 32511016-0 2020 Sunitinib inhibits PD-L1 expression in osteosarcoma by targeting STAT3 and remodels the immune system in tumor-bearing mice. Sunitinib 0-9 signal transducer and activator of transcription 3 Mus musculus 65-70 32511016-5 2020 Results: Sunitinib reduced the expression of PD-L1 by inhibiting the activation of STAT3 and suppressed the migration and invasion in osteosarcoma cells. Sunitinib 9-18 CD274 antigen Mus musculus 45-50 32511016-5 2020 Results: Sunitinib reduced the expression of PD-L1 by inhibiting the activation of STAT3 and suppressed the migration and invasion in osteosarcoma cells. Sunitinib 9-18 signal transducer and activator of transcription 3 Mus musculus 83-88 32511016-7 2020 Conclusion: Sunitinib inhibits PD-L1 expression by targeting STAT3 and remodels the immune system in tumor-bearing mice. Sunitinib 12-21 CD274 antigen Mus musculus 31-36 32511016-7 2020 Conclusion: Sunitinib inhibits PD-L1 expression by targeting STAT3 and remodels the immune system in tumor-bearing mice. Sunitinib 12-21 signal transducer and activator of transcription 3 Mus musculus 61-66 32358665-3 2020 Present hypothesis was aimed to examine the nephroprotective potential of RIVA in SUN-induced nephrotoxicity, mediated through the inhibition of oxidative stress-induced apoptosis and inflammation, via the TNF-alpha/NFk-B signaling pathways. Sunitinib 82-85 tumor necrosis factor Rattus norvegicus 206-215 32358665-3 2020 Present hypothesis was aimed to examine the nephroprotective potential of RIVA in SUN-induced nephrotoxicity, mediated through the inhibition of oxidative stress-induced apoptosis and inflammation, via the TNF-alpha/NFk-B signaling pathways. Sunitinib 82-85 nuclear factor kappa B subunit 1 Rattus norvegicus 216-221 32760216-3 2020 This study was assigned to probe the effect and mechanism of HOTAIR on sunitinib resistance of RC. Sunitinib 71-80 HOX transcript antisense RNA (non-protein coding) Mus musculus 61-67 32760216-11 2020 The role of HOTAIR knockdown in sunitinib resistance was verified in nude mice. Sunitinib 32-41 HOX transcript antisense RNA (non-protein coding) Mus musculus 12-18 32760216-12 2020 Results: HOTAIR expression in sunitinib-resistant cells is higher than that in parental cells. Sunitinib 30-39 HOX transcript antisense RNA (non-protein coding) Mus musculus 9-15 32760216-13 2020 Knockdown of HOTAIR in sunitinib-resistant cells lead to refrained sunitinib resistance and cell autophagy both in vivo and in vitro. Sunitinib 23-32 HOX transcript antisense RNA (non-protein coding) Mus musculus 13-19 32760216-13 2020 Knockdown of HOTAIR in sunitinib-resistant cells lead to refrained sunitinib resistance and cell autophagy both in vivo and in vitro. Sunitinib 67-76 HOX transcript antisense RNA (non-protein coding) Mus musculus 13-19 32760216-17 2020 Conclusion: HOTAIR negatively targets miR-17-5p to activate Beclin1-mediated cell autophagy, thereby enhancing sunitinib resistance in RC cells. Sunitinib 111-120 HOX transcript antisense RNA (non-protein coding) Mus musculus 12-18 32760216-17 2020 Conclusion: HOTAIR negatively targets miR-17-5p to activate Beclin1-mediated cell autophagy, thereby enhancing sunitinib resistance in RC cells. Sunitinib 111-120 beclin 1, autophagy related Mus musculus 60-67 32312893-0 2020 Identification of a RAS-activating TMEM87A-RASGRF1 fusion in an exceptional responder to sunitinib with non-small cell lung cancer. Sunitinib 89-98 transmembrane protein 87A Homo sapiens 35-42 32312893-0 2020 Identification of a RAS-activating TMEM87A-RASGRF1 fusion in an exceptional responder to sunitinib with non-small cell lung cancer. Sunitinib 89-98 Ras protein specific guanine nucleotide releasing factor 1 Homo sapiens 43-50 32469384-15 2020 Following availability of external data on PFS with sunitinib in patients with MET-driven disease, study enrollment was closed. Sunitinib 52-61 SAFB like transcription modulator Homo sapiens 79-82 32792963-7 2020 Moreover, we found that the levels of p-RB, CDK4, and Cyclin D1 were up-regulated in sunitinib resistant 786-O, OS-RC-2, and TK-10 cells, and inhibition of CDK4 by palbociclib or wogonin effectively reversed the sunitinib resistance, indicating that the hyperactivation of CDK4-RB pathway may at least partially contribute to the resistance of RCC to sunitinib. Sunitinib 85-94 RB transcriptional corepressor 1 Homo sapiens 38-42 32792963-7 2020 Moreover, we found that the levels of p-RB, CDK4, and Cyclin D1 were up-regulated in sunitinib resistant 786-O, OS-RC-2, and TK-10 cells, and inhibition of CDK4 by palbociclib or wogonin effectively reversed the sunitinib resistance, indicating that the hyperactivation of CDK4-RB pathway may at least partially contribute to the resistance of RCC to sunitinib. Sunitinib 85-94 cyclin dependent kinase 4 Homo sapiens 44-48 32792963-7 2020 Moreover, we found that the levels of p-RB, CDK4, and Cyclin D1 were up-regulated in sunitinib resistant 786-O, OS-RC-2, and TK-10 cells, and inhibition of CDK4 by palbociclib or wogonin effectively reversed the sunitinib resistance, indicating that the hyperactivation of CDK4-RB pathway may at least partially contribute to the resistance of RCC to sunitinib. Sunitinib 85-94 cyclin D1 Homo sapiens 54-63 32792963-7 2020 Moreover, we found that the levels of p-RB, CDK4, and Cyclin D1 were up-regulated in sunitinib resistant 786-O, OS-RC-2, and TK-10 cells, and inhibition of CDK4 by palbociclib or wogonin effectively reversed the sunitinib resistance, indicating that the hyperactivation of CDK4-RB pathway may at least partially contribute to the resistance of RCC to sunitinib. Sunitinib 212-221 RB transcriptional corepressor 1 Homo sapiens 38-42 32641718-4 2020 SNHG12 promoted RCC proliferation, migration, invasion and sunitinib resistance via CDCA3 in vitro. Sunitinib 59-68 small nucleolar RNA host gene 12 Homo sapiens 0-6 32792963-7 2020 Moreover, we found that the levels of p-RB, CDK4, and Cyclin D1 were up-regulated in sunitinib resistant 786-O, OS-RC-2, and TK-10 cells, and inhibition of CDK4 by palbociclib or wogonin effectively reversed the sunitinib resistance, indicating that the hyperactivation of CDK4-RB pathway may at least partially contribute to the resistance of RCC to sunitinib. Sunitinib 212-221 cyclin dependent kinase 4 Homo sapiens 44-48 32792963-7 2020 Moreover, we found that the levels of p-RB, CDK4, and Cyclin D1 were up-regulated in sunitinib resistant 786-O, OS-RC-2, and TK-10 cells, and inhibition of CDK4 by palbociclib or wogonin effectively reversed the sunitinib resistance, indicating that the hyperactivation of CDK4-RB pathway may at least partially contribute to the resistance of RCC to sunitinib. Sunitinib 212-221 cyclin D1 Homo sapiens 54-63 32792963-7 2020 Moreover, we found that the levels of p-RB, CDK4, and Cyclin D1 were up-regulated in sunitinib resistant 786-O, OS-RC-2, and TK-10 cells, and inhibition of CDK4 by palbociclib or wogonin effectively reversed the sunitinib resistance, indicating that the hyperactivation of CDK4-RB pathway may at least partially contribute to the resistance of RCC to sunitinib. Sunitinib 212-221 cyclin dependent kinase 4 Homo sapiens 156-160 32792963-7 2020 Moreover, we found that the levels of p-RB, CDK4, and Cyclin D1 were up-regulated in sunitinib resistant 786-O, OS-RC-2, and TK-10 cells, and inhibition of CDK4 by palbociclib or wogonin effectively reversed the sunitinib resistance, indicating that the hyperactivation of CDK4-RB pathway may at least partially contribute to the resistance of RCC to sunitinib. Sunitinib 212-221 cyclin dependent kinase 4 Homo sapiens 156-160 32792963-7 2020 Moreover, we found that the levels of p-RB, CDK4, and Cyclin D1 were up-regulated in sunitinib resistant 786-O, OS-RC-2, and TK-10 cells, and inhibition of CDK4 by palbociclib or wogonin effectively reversed the sunitinib resistance, indicating that the hyperactivation of CDK4-RB pathway may at least partially contribute to the resistance of RCC to sunitinib. Sunitinib 212-221 RB transcriptional corepressor 1 Homo sapiens 38-42 32792963-7 2020 Moreover, we found that the levels of p-RB, CDK4, and Cyclin D1 were up-regulated in sunitinib resistant 786-O, OS-RC-2, and TK-10 cells, and inhibition of CDK4 by palbociclib or wogonin effectively reversed the sunitinib resistance, indicating that the hyperactivation of CDK4-RB pathway may at least partially contribute to the resistance of RCC to sunitinib. Sunitinib 212-221 cyclin dependent kinase 4 Homo sapiens 44-48 32792963-7 2020 Moreover, we found that the levels of p-RB, CDK4, and Cyclin D1 were up-regulated in sunitinib resistant 786-O, OS-RC-2, and TK-10 cells, and inhibition of CDK4 by palbociclib or wogonin effectively reversed the sunitinib resistance, indicating that the hyperactivation of CDK4-RB pathway may at least partially contribute to the resistance of RCC to sunitinib. Sunitinib 212-221 cyclin D1 Homo sapiens 54-63 32792963-7 2020 Moreover, we found that the levels of p-RB, CDK4, and Cyclin D1 were up-regulated in sunitinib resistant 786-O, OS-RC-2, and TK-10 cells, and inhibition of CDK4 by palbociclib or wogonin effectively reversed the sunitinib resistance, indicating that the hyperactivation of CDK4-RB pathway may at least partially contribute to the resistance of RCC to sunitinib. Sunitinib 212-221 cyclin dependent kinase 4 Homo sapiens 156-160 32792963-7 2020 Moreover, we found that the levels of p-RB, CDK4, and Cyclin D1 were up-regulated in sunitinib resistant 786-O, OS-RC-2, and TK-10 cells, and inhibition of CDK4 by palbociclib or wogonin effectively reversed the sunitinib resistance, indicating that the hyperactivation of CDK4-RB pathway may at least partially contribute to the resistance of RCC to sunitinib. Sunitinib 212-221 cyclin dependent kinase 4 Homo sapiens 156-160 32698382-8 2020 Furthermore, a decrease in nitric oxide (NO) levels and endothelial nitric oxide synthase (eNOS) activation was observed in aortas from Su-treated animals. Sunitinib 136-138 nitric oxide synthase 3 Rattus norvegicus 56-89 32698382-8 2020 Furthermore, a decrease in nitric oxide (NO) levels and endothelial nitric oxide synthase (eNOS) activation was observed in aortas from Su-treated animals. Sunitinib 136-138 nitric oxide synthase 3 Rattus norvegicus 91-95 32234492-8 2020 Low expression of JAK2 were resistant to QL-VIII-58, TL-1-85, Ruxolitinib, TG101348 and Sunitinib. Sunitinib 88-97 Janus kinase 2 Homo sapiens 18-22 32641718-0 2020 Long noncoding RNA SNHG12 promotes tumour progression and sunitinib resistance by upregulating CDCA3 in renal cell carcinoma. Sunitinib 58-67 small nucleolar RNA host gene 12 Homo sapiens 19-25 32641718-2 2020 Long noncoding RNA SNHG12 (lncRNA SNHG12) has emerged as a key molecule in numerous human cancers, but its functions in renal cell carcinoma (RCC) sunitinib resistance remain unclear. Sunitinib 147-156 small nucleolar RNA host gene 12 Homo sapiens 19-25 32641718-2 2020 Long noncoding RNA SNHG12 (lncRNA SNHG12) has emerged as a key molecule in numerous human cancers, but its functions in renal cell carcinoma (RCC) sunitinib resistance remain unclear. Sunitinib 147-156 small nucleolar RNA host gene 12 Homo sapiens 34-40 32641718-3 2020 In this study, we found SNHG12 was highly expressed in RCC tissues and in sunitinib-resistant RCC cells and was associated with a poor clinical prognosis. Sunitinib 74-83 small nucleolar RNA host gene 12 Homo sapiens 24-30 32667929-7 2020 In RCC cells from mice treated with disulfiram and/or sunitinib, several genes associated with serine biosynthesis and aldose reductase were downregulated in cells treated with disulfiram or sunitinib alone and further downregulated in cells treated with both disulfiram and sunitinib. Sunitinib 54-63 aldo-keto reductase family 1, member B3 (aldose reductase) Mus musculus 119-135 32667929-7 2020 In RCC cells from mice treated with disulfiram and/or sunitinib, several genes associated with serine biosynthesis and aldose reductase were downregulated in cells treated with disulfiram or sunitinib alone and further downregulated in cells treated with both disulfiram and sunitinib. Sunitinib 191-200 aldo-keto reductase family 1, member B3 (aldose reductase) Mus musculus 119-135 32667929-7 2020 In RCC cells from mice treated with disulfiram and/or sunitinib, several genes associated with serine biosynthesis and aldose reductase were downregulated in cells treated with disulfiram or sunitinib alone and further downregulated in cells treated with both disulfiram and sunitinib. Sunitinib 191-200 aldo-keto reductase family 1, member B3 (aldose reductase) Mus musculus 119-135 32802186-14 2020 Moreover, silencing of GYS1 increased the synthetic lethality of ccRCC cells to sunitinib treatment by concomitantly suppressing p65. Sunitinib 80-89 glycogen synthase 1 Homo sapiens 23-27 32802186-16 2020 Re-sensitization of ccRCC cells to sunitinib suggests that GYS1 is a useful indicator of unfavorable prognosis as well as a therapeutic target for patients with ccRCC. Sunitinib 35-44 glycogen synthase 1 Homo sapiens 59-63 32641718-7 2020 Furthermore, in vivo experiments showed that SNHG12 increased tumour growth and that knocking down SNHG12 could reverse RCC sunitinib resistance. Sunitinib 124-133 small nucleolar RNA host gene 12 Homo sapiens 99-105 32641718-8 2020 Our study revealed that the lncRNA SNHG12/SP1/CDCA3 axis promoted RCC progression and sunitinib resistance, which could provide a new therapeutic target for sunitinib-resistant RCC. Sunitinib 86-95 small nucleolar RNA host gene 12 Homo sapiens 35-41 32641718-8 2020 Our study revealed that the lncRNA SNHG12/SP1/CDCA3 axis promoted RCC progression and sunitinib resistance, which could provide a new therapeutic target for sunitinib-resistant RCC. Sunitinib 86-95 cell division cycle associated 3 Homo sapiens 46-51 32641718-8 2020 Our study revealed that the lncRNA SNHG12/SP1/CDCA3 axis promoted RCC progression and sunitinib resistance, which could provide a new therapeutic target for sunitinib-resistant RCC. Sunitinib 157-166 small nucleolar RNA host gene 12 Homo sapiens 35-41 32641718-8 2020 Our study revealed that the lncRNA SNHG12/SP1/CDCA3 axis promoted RCC progression and sunitinib resistance, which could provide a new therapeutic target for sunitinib-resistant RCC. Sunitinib 157-166 cell division cycle associated 3 Homo sapiens 46-51 32233096-5 2020 Through screening, we identified two FDA-approved drugs, Oxytetracycline and Sunitinib, which were able to dissociate Abeta oligomers and plaques to monomers in 5XFAD transgenic mice. Sunitinib 77-86 amyloid beta (A4) precursor protein Mus musculus 118-123 32334266-4 2020 The multi-kinase inhibitor, sunitinib, has demonstrated potent nanomolar inhibition of AMPK activity and has scope for modification. Sunitinib 28-37 protein kinase AMP-activated catalytic subunit alpha 2 Homo sapiens 87-91 32126313-6 2020 Sunitinib, a tyrosine receptor kinase inhibitor that also blocks VEGFR2 blocked sildenafil-/vardenafil-induced osteoblast differentiation. Sunitinib 0-9 kinase insert domain protein receptor Mus musculus 65-71 32334266-6 2020 We identified two potent, novel oxindole-based AMPK inhibitors that were designed to interact with the DFG motif in the ATP-binding site of AMPK, this key feature evades interaction with the common recptor tyrosine kinase targets of sunitinib. Sunitinib 233-242 protein kinase AMP-activated catalytic subunit alpha 2 Homo sapiens 47-51 32334266-6 2020 We identified two potent, novel oxindole-based AMPK inhibitors that were designed to interact with the DFG motif in the ATP-binding site of AMPK, this key feature evades interaction with the common recptor tyrosine kinase targets of sunitinib. Sunitinib 233-242 protein kinase AMP-activated catalytic subunit alpha 2 Homo sapiens 140-144 32661119-8 2020 The most commonly used IO and VEGF-TKIs were nivolumab (66%) and sunitinib (40%). Sunitinib 65-74 vascular endothelial growth factor A Homo sapiens 30-34 32382017-0 2020 ZHX2 drives cell growth and migration via activating MEK/ERK signal and induces Sunitinib resistance by regulating the autophagy in clear cell Renal Cell Carcinoma. Sunitinib 80-89 zinc fingers and homeoboxes 2 Homo sapiens 0-4 32409702-0 2020 Targeting the ERbeta/Angiopoietin-2/Tie-2 signaling-mediated angiogenesis with the FDA-approved anti-estrogen Faslodex to increase the Sunitinib sensitivity in RCC. Sunitinib 135-144 estrogen receptor 1 (alpha) Mus musculus 14-20 32409702-0 2020 Targeting the ERbeta/Angiopoietin-2/Tie-2 signaling-mediated angiogenesis with the FDA-approved anti-estrogen Faslodex to increase the Sunitinib sensitivity in RCC. Sunitinib 135-144 angiopoietin 2 Mus musculus 21-35 32409702-0 2020 Targeting the ERbeta/Angiopoietin-2/Tie-2 signaling-mediated angiogenesis with the FDA-approved anti-estrogen Faslodex to increase the Sunitinib sensitivity in RCC. Sunitinib 135-144 TEK receptor tyrosine kinase Mus musculus 36-41 32409702-6 2020 Our study is the first to explore (i) how estrogen receptor beta (ERbeta) can up-regulate ANGPT-2 in RCC cells, and (ii) how ERbeta-increased ANGPT-2 can promote the HUVEC tube formation and reduce sunitinib sensitivity. Sunitinib 198-207 estrogen receptor 1 (alpha) Mus musculus 66-72 32409702-6 2020 Our study is the first to explore (i) how estrogen receptor beta (ERbeta) can up-regulate ANGPT-2 in RCC cells, and (ii) how ERbeta-increased ANGPT-2 can promote the HUVEC tube formation and reduce sunitinib sensitivity. Sunitinib 198-207 estrogen receptor 1 (alpha) Mus musculus 125-131 32409702-6 2020 Our study is the first to explore (i) how estrogen receptor beta (ERbeta) can up-regulate ANGPT-2 in RCC cells, and (ii) how ERbeta-increased ANGPT-2 can promote the HUVEC tube formation and reduce sunitinib sensitivity. Sunitinib 198-207 angiopoietin 2 Mus musculus 142-149 32409702-8 2020 We found the up-regulated ANGPT-2 of RCC cells could then increase the Tie-2 phosphorylation to promote the angiogenesis and increase sunitinib treatment resistance of endothelial cells. Sunitinib 134-143 angiopoietin 2 Mus musculus 26-33 32409702-8 2020 We found the up-regulated ANGPT-2 of RCC cells could then increase the Tie-2 phosphorylation to promote the angiogenesis and increase sunitinib treatment resistance of endothelial cells. Sunitinib 134-143 TEK receptor tyrosine kinase Mus musculus 71-76 32409702-10 2020 Targeting this newly identified ERbeta/ANGPT-2/Tie-2 signaling pathway with the FDA-approved anti-estrogen, Faslodex, may help in the development of a novel combined therapy with sunitinib to better suppress the ccRCC progression. Sunitinib 179-188 estrogen receptor 1 (alpha) Mus musculus 32-38 32409702-10 2020 Targeting this newly identified ERbeta/ANGPT-2/Tie-2 signaling pathway with the FDA-approved anti-estrogen, Faslodex, may help in the development of a novel combined therapy with sunitinib to better suppress the ccRCC progression. Sunitinib 179-188 angiopoietin 2 Mus musculus 39-46 32409702-10 2020 Targeting this newly identified ERbeta/ANGPT-2/Tie-2 signaling pathway with the FDA-approved anti-estrogen, Faslodex, may help in the development of a novel combined therapy with sunitinib to better suppress the ccRCC progression. Sunitinib 179-188 TEK receptor tyrosine kinase Mus musculus 47-52 32382017-9 2020 We also found ZHX2 overexpression induce Sunitinib resistance though activating autophagy and the combination treatment of Sunitinib and Chloroquine could significantly rescue the phenomenon. Sunitinib 41-50 zinc fingers and homeoboxes 2 Homo sapiens 14-18 32382017-9 2020 We also found ZHX2 overexpression induce Sunitinib resistance though activating autophagy and the combination treatment of Sunitinib and Chloroquine could significantly rescue the phenomenon. Sunitinib 123-132 zinc fingers and homeoboxes 2 Homo sapiens 14-18 32382017-10 2020 In summary, these results indicate that ZHX2 drivers cell growth, migration though increase VEGF expression, and transcriptional activate MEK/ERK1/2 signaling pathway, and could induce Sunitinib resistance by regulating self-protective autophagy, these may provide new insight in advanced ccRCC treatment. Sunitinib 185-194 zinc fingers and homeoboxes 2 Homo sapiens 40-44 32379831-0 2020 Oncogenic effects of RAB27B through exosome independent function in renal cell carcinoma including sunitinib-resistant. Sunitinib 99-108 RAB27B, member RAS oncogene family Homo sapiens 21-27 32232883-0 2020 Potential new therapy of Rapalink-1, a new generation mTOR inhibitor, against sunitinib-resistant renal cell carcinoma. Sunitinib 78-87 mechanistic target of rapamycin kinase Homo sapiens 54-58 32379831-7 2020 The present study aimed to elucidate the role of RAB27B in RCC including sunitinib-resistant and its role in exosomes. Sunitinib 73-82 RAB27B, member RAS oncogene family Homo sapiens 49-55 32379831-10 2020 Furthermore, RAB27B protein expression was enhanced in all of the tested sunitinib-resistant RCC cell lines compared to parental cells. Sunitinib 73-82 RAB27B, member RAS oncogene family Homo sapiens 13-19 32379831-12 2020 Moreover, anti-tumor effects of RAB27B downregulation were also observed in sunitinib-resistant RCC cells. Sunitinib 76-85 RAB27B, member RAS oncogene family Homo sapiens 32-38 32379831-14 2020 These results showed that RAB27B is a prognostic marker and a novel therapeutic target in sunitinib-sensitive and -resistant RCCs. Sunitinib 90-99 RAB27B, member RAS oncogene family Homo sapiens 26-32 32232883-1 2020 Sunitinib, a muiti-targeted receptor tyrosine kinase inhibitor including vascular endothelial growth factor, has been widely used as a first-line treatment against metastatic renal cell carcinoma (mRCC). Sunitinib 0-9 vascular endothelial growth factor A Homo sapiens 73-107 32272660-0 2020 The Efficacy of Sunitinib Treatment of Renal Cancer Cells Is Associated with the Protein PHAX In Vitro. Sunitinib 16-25 phosphorylated adaptor for RNA export Homo sapiens 89-93 32355171-11 2020 There was a significantly lower expression of both VEGFR-1 and FSHR molecular ultrasound imaging signals in the sunitinib group at all times of treatment (Days 7, 14 and 28). Sunitinib 112-121 fms related receptor tyrosine kinase 1 Homo sapiens 51-58 32355171-11 2020 There was a significantly lower expression of both VEGFR-1 and FSHR molecular ultrasound imaging signals in the sunitinib group at all times of treatment (Days 7, 14 and 28). Sunitinib 112-121 follicle stimulating hormone receptor Homo sapiens 63-67 32355171-14 2020 This study demonstrated for the first time the potential of VEGFR1 and FSHR, by ultrasound-based molecular imaging, to follow-up the longitudinal response to sunitinib in ccRCC. Sunitinib 158-167 fms related receptor tyrosine kinase 1 Homo sapiens 60-66 32355171-14 2020 This study demonstrated for the first time the potential of VEGFR1 and FSHR, by ultrasound-based molecular imaging, to follow-up the longitudinal response to sunitinib in ccRCC. Sunitinib 158-167 follicle stimulating hormone receptor Homo sapiens 71-75 32321460-0 2020 Tumor endothelial ELTD1 as a predictive marker for treatment of renal cancer patients with sunitinib. Sunitinib 91-100 adhesion G protein-coupled receptor L4 Homo sapiens 18-23 32321460-9 2020 RESULTS: Patients with high ELTD1 expression in the tumor vasculature experienced a significantly better progression free survival (PFS) with sunitinib treatment as compared to patients with low ELTD1 expression (8 versus 5.5 months, respectively). Sunitinib 142-151 adhesion G protein-coupled receptor L4 Homo sapiens 28-33 32321460-12 2020 CONCLUSIONS: Our results identify tumor vessel ELTD1 expression as a positive predictive marker for sunitinib-treatment in patients suffering from mRCC. Sunitinib 100-109 adhesion G protein-coupled receptor L4 Homo sapiens 47-52 32272660-2 2020 Here, we take steps towards validating a computational prediction based on differential transcriptome network analysis that phosphorylated adapter RNA export protein (PHAX) is associated with sunitinib drug treatment. Sunitinib 192-201 phosphorylated adaptor for RNA export Homo sapiens 124-165 32272660-2 2020 Here, we take steps towards validating a computational prediction based on differential transcriptome network analysis that phosphorylated adapter RNA export protein (PHAX) is associated with sunitinib drug treatment. Sunitinib 192-201 phosphorylated adaptor for RNA export Homo sapiens 167-171 32272660-4 2020 Immunofluorescence staining of patient tumours showed strong localisation of PHAX to the microvasculature consistent with the anti-angiogenic effect of sunitinib. Sunitinib 152-161 phosphorylated adaptor for RNA export Homo sapiens 77-81 32272660-6 2020 In organ culture, ccRCC cells had higher levels of PHAX protein expression than normal kidney cells, and sunitinib increased PHAX protein expression in a dose dependent manner (untreated vs. 100 microM; p < 0.05). Sunitinib 105-114 phosphorylated adaptor for RNA export Homo sapiens 125-129 32272660-7 2020 PHAX knockdown in a ccRCC organ culture model impacted the ability of sunitinib to cause cancer cell death (p < 0.0001 untreated vs. treated), suggesting a role for PHAX in mediating the efficacy of sunitinib. Sunitinib 70-79 phosphorylated adaptor for RNA export Homo sapiens 0-4 32272660-7 2020 PHAX knockdown in a ccRCC organ culture model impacted the ability of sunitinib to cause cancer cell death (p < 0.0001 untreated vs. treated), suggesting a role for PHAX in mediating the efficacy of sunitinib. Sunitinib 199-208 phosphorylated adaptor for RNA export Homo sapiens 0-4 32272660-7 2020 PHAX knockdown in a ccRCC organ culture model impacted the ability of sunitinib to cause cancer cell death (p < 0.0001 untreated vs. treated), suggesting a role for PHAX in mediating the efficacy of sunitinib. Sunitinib 199-208 phosphorylated adaptor for RNA export Homo sapiens 165-169 31369705-4 2020 Interestingly, our data indicated that one candidate aptamer, called V15, can specifically inhibit the in vitro kinase activity of mutant c-KITD816V with an IC50 value that is 9-fold more potent than the sunitinib drug tested under the same conditions. Sunitinib 204-213 immunoglobulin lambda variable 2-18 Homo sapiens 69-72 31999483-9 2020 Besides, we found that sunitinib, a multi-targeted tyrosine kinase inhibitor, could significantly inhibit ufmylation, whereas overexpression of active Ufm1 partially inhibited the antitumor effects of sunitinib. Sunitinib 201-210 ubiquitin fold modifier 1 Homo sapiens 151-155 31866228-3 2020 Another class of therapeutic agents that has been implicated in TMA is the vascular endothelial growth factor (VEGF) pathway inhibitors, including the anti-VEGF monoclonal antibody bevacizumab and the VEGF receptor tyrosine kinase inhibitor sunitinib. Sunitinib 241-250 vascular endothelial growth factor A Homo sapiens 75-109 31866228-3 2020 Another class of therapeutic agents that has been implicated in TMA is the vascular endothelial growth factor (VEGF) pathway inhibitors, including the anti-VEGF monoclonal antibody bevacizumab and the VEGF receptor tyrosine kinase inhibitor sunitinib. Sunitinib 241-250 vascular endothelial growth factor A Homo sapiens 111-115 32234883-0 2020 PTEN Is Involved in Sunitinib and Sorafenib Resistance in Renal Cell Carcinoma. Sunitinib 20-29 phosphatase and tensin homolog Homo sapiens 0-4 32234883-5 2020 RESULTS: PTEN knockout promoted spheroid formation and decreased sunitinib/sorafenib sensitivity in RCC cell lines. Sunitinib 65-74 phosphatase and tensin homolog Homo sapiens 9-13 32234883-6 2020 PTEN immunohistochemistry in 74 metastatic RCCs treated with sunitinib and sorafenib revealed negative PTEN expression in 23% of samples. Sunitinib 61-70 phosphatase and tensin homolog Homo sapiens 0-4 32234883-7 2020 Kaplan-Meier analysis showed a significant association of negative PTEN expression with poor progression-free survival in metastatic RCC treated with sunitinib and sorafenib (p=0.024) or sunitinib alone (p=0.009). Sunitinib 150-159 phosphatase and tensin homolog Homo sapiens 67-71 32234883-7 2020 Kaplan-Meier analysis showed a significant association of negative PTEN expression with poor progression-free survival in metastatic RCC treated with sunitinib and sorafenib (p=0.024) or sunitinib alone (p=0.009). Sunitinib 187-196 phosphatase and tensin homolog Homo sapiens 67-71 32078941-6 2020 Sunitinib malate increased infiltration of CD8+ T cells and decreased regulatory T cells (Tregs), accompanied by inhibited expression of TGF-beta1 and IL-10 and increased CCL-28, IFN-gamma and IL-12, but no significant inhibition of myeloid-derived suppressor cells (MDSCs) was observed. Sunitinib 0-16 transforming growth factor beta 1 Homo sapiens 137-146 32078941-6 2020 Sunitinib malate increased infiltration of CD8+ T cells and decreased regulatory T cells (Tregs), accompanied by inhibited expression of TGF-beta1 and IL-10 and increased CCL-28, IFN-gamma and IL-12, but no significant inhibition of myeloid-derived suppressor cells (MDSCs) was observed. Sunitinib 0-16 interleukin 10 Homo sapiens 151-156 32078941-6 2020 Sunitinib malate increased infiltration of CD8+ T cells and decreased regulatory T cells (Tregs), accompanied by inhibited expression of TGF-beta1 and IL-10 and increased CCL-28, IFN-gamma and IL-12, but no significant inhibition of myeloid-derived suppressor cells (MDSCs) was observed. Sunitinib 0-16 C-C motif chemokine ligand 28 Homo sapiens 171-177 32078941-6 2020 Sunitinib malate increased infiltration of CD8+ T cells and decreased regulatory T cells (Tregs), accompanied by inhibited expression of TGF-beta1 and IL-10 and increased CCL-28, IFN-gamma and IL-12, but no significant inhibition of myeloid-derived suppressor cells (MDSCs) was observed. Sunitinib 0-16 interferon gamma Homo sapiens 179-188 32078941-7 2020 In addition, sunitinib malate increased the levels of PD-1 and PD-L1 in TME, combined with anti-PD-1 therapy showed a significant reduction in tumor burden compared with either monotherapy, suggesting that anti-PD-1 therapy is reasonable after sunitinib malate treatment. Sunitinib 13-29 CD274 molecule Homo sapiens 63-68 32078941-7 2020 In addition, sunitinib malate increased the levels of PD-1 and PD-L1 in TME, combined with anti-PD-1 therapy showed a significant reduction in tumor burden compared with either monotherapy, suggesting that anti-PD-1 therapy is reasonable after sunitinib malate treatment. Sunitinib 244-260 CD274 molecule Homo sapiens 63-68 31702819-3 2020 Previous reports have found success with sunitinib in imatinib-resistant GIST, but we report a certain wild-type KIT mutation GIST with cutaneous and subcutaneous metastasis that was unresponsive to multiple tyrosine kinase inhibitor (TKI) treatments. Sunitinib 41-50 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 113-116 32041631-8 2020 RESULTS: We demonstrate that YB-1 and ABCB-1 are upregulated in sunitinib-resistant in vitro, ex vivo, in vivo and patient samples compared to the sensitive samples. Sunitinib 64-73 Y-box binding protein 1 Homo sapiens 29-33 31023123-0 2020 Influence of ABCB1 (1236C > T, 2677G > T and 3435C > T) polymorphisms on the transport ability of P-gp-mediated sunitinib in Caco-2 cell line. Sunitinib 121-130 ATP binding cassette subfamily B member 1 Homo sapiens 13-18 31023123-0 2020 Influence of ABCB1 (1236C > T, 2677G > T and 3435C > T) polymorphisms on the transport ability of P-gp-mediated sunitinib in Caco-2 cell line. Sunitinib 121-130 ATP binding cassette subfamily B member 1 Homo sapiens 107-111 31023123-2 2020 The aim of the current research was to observe in vitro the impacts of ABCB1 (1236 C > T, 2677G > T, and 3435C > T) polymorphisms on the efflux activity of P-gp-mediated sunitinib. Sunitinib 179-188 ATP binding cassette subfamily B member 1 Homo sapiens 71-76 31023123-2 2020 The aim of the current research was to observe in vitro the impacts of ABCB1 (1236 C > T, 2677G > T, and 3435C > T) polymorphisms on the efflux activity of P-gp-mediated sunitinib. Sunitinib 179-188 ATP binding cassette subfamily B member 1 Homo sapiens 165-169 31023123-6 2020 The recombinant cell lines transfected with ABCB1 variant alleles (1236 T, 2677T, and 3435T) showed higher resistance to sunitinib compared to cells transfecting with ABCB1 wild-type allele (p < .05). Sunitinib 121-130 ATP binding cassette subfamily B member 1 Homo sapiens 44-49 31023123-7 2020 The intracellular accumulation of sunitinib was significantly decreased in the three types of recombinant cell lines overexpressing ABCB1 variant alleles in comparison of their wild-type cell lines (p < .05). Sunitinib 34-43 ATP binding cassette subfamily B member 1 Homo sapiens 132-137 31023123-9 2020 The P-gp activity in recombinant variant cells is stronger when mediated transport of sunitinib than wild-type counterpart. Sunitinib 86-95 ATP binding cassette subfamily B member 1 Homo sapiens 4-8 31023123-10 2020 P-gp encoded by ABCB1 (1236 T, 2677T, and 3435T) variant alleles may be more efficient to transport sunitinib than wild-type allele. Sunitinib 100-109 ATP binding cassette subfamily B member 1 Homo sapiens 0-4 31023123-10 2020 P-gp encoded by ABCB1 (1236 T, 2677T, and 3435T) variant alleles may be more efficient to transport sunitinib than wild-type allele. Sunitinib 100-109 ATP binding cassette subfamily B member 1 Homo sapiens 16-21 31023123-11 2020 Our observation suggests that ABCB1 (1236 C > T, 2677G > T, and 3435C > T) polymorphisms affect the transport ability of P-gp-mediated sunitinib. Sunitinib 144-153 ATP binding cassette subfamily B member 1 Homo sapiens 30-35 31023123-11 2020 Our observation suggests that ABCB1 (1236 C > T, 2677G > T, and 3435C > T) polymorphisms affect the transport ability of P-gp-mediated sunitinib. Sunitinib 144-153 ATP binding cassette subfamily B member 1 Homo sapiens 130-134 31023123-12 2020 Collectively, ABCB1 polymorphisms may alter the P-gp-mediated sunitinib sensitivity via regulating drug transport. Sunitinib 62-71 ATP binding cassette subfamily B member 1 Homo sapiens 14-19 31023123-12 2020 Collectively, ABCB1 polymorphisms may alter the P-gp-mediated sunitinib sensitivity via regulating drug transport. Sunitinib 62-71 ATP binding cassette subfamily B member 1 Homo sapiens 48-52 32041631-8 2020 RESULTS: We demonstrate that YB-1 and ABCB-1 are upregulated in sunitinib-resistant in vitro, ex vivo, in vivo and patient samples compared to the sensitive samples. Sunitinib 64-73 ATP binding cassette subfamily B member 1 Homo sapiens 38-44 32041631-10 2020 Furthermore, our results establish that inhibiting ABCB-1 with elacridar, in addition to sunitinib, has a positive impact on reverting sunitinib-resistance development in in vitro, ex vivo and in vivo models. Sunitinib 135-144 ATP binding cassette subfamily B member 1 Homo sapiens 51-57 31922424-3 2020 In November 2017, sunitinib was approved in the USA as the first adjuvant therapy for patients at high risk for recurrent RCC postnephrectomy based on results from the S-TRAC trial. Sunitinib 18-27 T cell receptor alpha constant Homo sapiens 170-174 32206105-2 2020 Using a new multimodal preclinical imaging instrument, we explored a sequence of events leading to sunitinib-induced resistance in a murine model of paraganglioma (PGL) invalidated for the expression of succinate dehydrogenase subunit B (Sdhb -/-). Sunitinib 99-108 succinate dehydrogenase complex, subunit B, iron sulfur (Ip) Mus musculus 238-242 32206105-10 2020 Conclusion: These results demonstrate an adaptive resistance of Sdhb -/- tumors to six weeks of sunitinib treatment. Sunitinib 96-105 succinate dehydrogenase complex, subunit B, iron sulfur (Ip) Mus musculus 64-68 31789391-7 2020 The results of western blotting demonstrated that sunitinib induced upregulation of LAMP-1 and LC3-II, and downregulation of sequestosome 1/p62, indicating the activation of autophagy. Sunitinib 50-59 lysosomal associated membrane protein 1 Homo sapiens 84-90 32005261-2 2020 The events that drive GIST oncogenesis are primarily KIT or PDGFRA mutations, which lead to the susceptibility of these tumors to small-molecule tyrosine kinase inhibitors such as imatinib and sunitinib. Sunitinib 193-202 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 53-56 32005261-2 2020 The events that drive GIST oncogenesis are primarily KIT or PDGFRA mutations, which lead to the susceptibility of these tumors to small-molecule tyrosine kinase inhibitors such as imatinib and sunitinib. Sunitinib 193-202 platelet derived growth factor receptor alpha Homo sapiens 60-66 31975150-4 2020 First, the PBPK model for sunitinib was established, then the cytochrome P450 3A4-mediated metabolism of sunitinib was used as the input for SU012662. Sunitinib 105-114 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 62-81 30825427-8 2020 Sunitinib malate, a multi tyrosine kinase inhibitor, market authorised for cancer, reversed the ocular behavioural and morphological phenotypes observed in vhl-/- zebrafish. Sunitinib 0-16 von Hippel-Lindau tumor suppressor Danio rerio 156-159 30825427-9 2020 We conclude that the zebrafish vhl gene contributes to an endogenous molecular barrier that prevents development of intraretinal vasculature, and that pharmacological intervention with sunitinib can improve visual function and hyaloid vessel patterning while reducing abnormally formed ectopic intraretinal vessels in vhl-/- zebrafish. Sunitinib 185-194 von Hippel-Lindau tumor suppressor Danio rerio 31-34 30825427-9 2020 We conclude that the zebrafish vhl gene contributes to an endogenous molecular barrier that prevents development of intraretinal vasculature, and that pharmacological intervention with sunitinib can improve visual function and hyaloid vessel patterning while reducing abnormally formed ectopic intraretinal vessels in vhl-/- zebrafish. Sunitinib 185-194 von Hippel-Lindau tumor suppressor Danio rerio 318-321 31744648-1 2020 INTRODUCTION: Sunitinib (SUN) and pazopanib (PAZ) are 2 oral tyrosine kinase inhibitors against vascular endothelial growth factor. Sunitinib 14-23 vascular endothelial growth factor A Homo sapiens 96-130 31535851-7 2020 Docking experiments in the CYP3A4 active site were performed and showed that pazopanib and sunitinib fitting in the catalytic site are in accordance with their regioselective oxidation to aldehydes. Sunitinib 91-100 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 27-33 31789391-7 2020 The results of western blotting demonstrated that sunitinib induced upregulation of LAMP-1 and LC3-II, and downregulation of sequestosome 1/p62, indicating the activation of autophagy. Sunitinib 50-59 microtubule associated protein 1 light chain 3 alpha Homo sapiens 95-98 31789391-7 2020 The results of western blotting demonstrated that sunitinib induced upregulation of LAMP-1 and LC3-II, and downregulation of sequestosome 1/p62, indicating the activation of autophagy. Sunitinib 50-59 sequestosome 1 Homo sapiens 125-139 31789391-7 2020 The results of western blotting demonstrated that sunitinib induced upregulation of LAMP-1 and LC3-II, and downregulation of sequestosome 1/p62, indicating the activation of autophagy. Sunitinib 50-59 sequestosome 1 Homo sapiens 140-143 31371341-0 2019 PD-L1 Expression and Clinical Outcomes to Cabozantinib, Everolimus, and Sunitinib in Patients with Metastatic Renal Cell Carcinoma: Analysis of the Randomized Clinical Trials METEOR and CABOSUN. Sunitinib 72-81 CD274 molecule Homo sapiens 0-5 31905947-6 2019 Inhibition of AAK1 kinase activity by sunitinib blocked AP2M1 phosphorylation, significantly inhibiting RABV infection and preventing RABV from entering early endosomes. Sunitinib 38-47 AP2 associated kinase 1 Homo sapiens 14-18 31905947-6 2019 Inhibition of AAK1 kinase activity by sunitinib blocked AP2M1 phosphorylation, significantly inhibiting RABV infection and preventing RABV from entering early endosomes. Sunitinib 38-47 adaptor related protein complex 2 subunit mu 1 Homo sapiens 56-61 31471313-2 2019 Approved KIT inhibitors sunitinib and regorafenib have complementary activity against KIT resistance mutations. Sunitinib 24-33 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 9-12 31471313-2 2019 Approved KIT inhibitors sunitinib and regorafenib have complementary activity against KIT resistance mutations. Sunitinib 24-33 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 86-89 31463627-6 2019 Sunitinib also significantly decreased the number of tumor-infiltrating CD8 + T and B cells, MDSCs, and macrophages. Sunitinib 0-9 CD8a molecule Homo sapiens 72-75 31485624-10 2019 GNF-5837 also enhanced the cytotoxic effects of sunitinib via inhibition of ERK kinase. Sunitinib 48-57 mitogen-activated protein kinase 1 Homo sapiens 76-79 31501521-0 2020 Targeting the TR4 nuclear receptor-mediated lncTASR/AXL signaling with tretinoin increases the sunitinib sensitivity to better suppress the RCC progression. Sunitinib 95-104 nuclear receptor subfamily 2 group C member 2 Homo sapiens 14-17 31501521-0 2020 Targeting the TR4 nuclear receptor-mediated lncTASR/AXL signaling with tretinoin increases the sunitinib sensitivity to better suppress the RCC progression. Sunitinib 95-104 AXL receptor tyrosine kinase Homo sapiens 52-55 31501521-4 2020 Here we found TR4 nuclear receptor might alter the sunitinib resistance to RCC via altering the TR4/lncTASR/AXL signaling. Sunitinib 51-60 nuclear receptor subfamily 2 group C member 2 Homo sapiens 14-17 31501521-4 2020 Here we found TR4 nuclear receptor might alter the sunitinib resistance to RCC via altering the TR4/lncTASR/AXL signaling. Sunitinib 51-60 nuclear receptor subfamily 2 group C member 2 Homo sapiens 96-99 31501521-4 2020 Here we found TR4 nuclear receptor might alter the sunitinib resistance to RCC via altering the TR4/lncTASR/AXL signaling. Sunitinib 51-60 AXL receptor tyrosine kinase Homo sapiens 108-111 31501521-5 2020 Mechanism dissection revealed that TR4 could modulate lncTASR (ENST00000600671.1) expression via transcriptional regulation, which might then increase AXL protein expression via enhancing the stability of AXL mRNA to increase the sunitinib resistance in RCC. Sunitinib 230-239 nuclear receptor subfamily 2 group C member 2 Homo sapiens 35-38 31501521-6 2020 Human clinical surveys also linked the expression of TR4, lncTASR, and AXL to the RCC survival, and results from multiple RCC cell lines revealed that targeting this newly identified TR4-mediated signaling with small molecules, including tretinoin, metformin, or TR4-shRNAs, all led to increase the sunitinib sensitivity to better suppress the RCC progression, and our preclinical study using the in vivo mouse model further proved tretinoin had a better synergistic effect to increase sunitinib sensitivity to suppress RCC progression. Sunitinib 299-308 nuclear receptor subfamily 2 group C member 2 Homo sapiens 183-186 31501521-6 2020 Human clinical surveys also linked the expression of TR4, lncTASR, and AXL to the RCC survival, and results from multiple RCC cell lines revealed that targeting this newly identified TR4-mediated signaling with small molecules, including tretinoin, metformin, or TR4-shRNAs, all led to increase the sunitinib sensitivity to better suppress the RCC progression, and our preclinical study using the in vivo mouse model further proved tretinoin had a better synergistic effect to increase sunitinib sensitivity to suppress RCC progression. Sunitinib 299-308 nuclear receptor subfamily 2 group C member 2 Homo sapiens 183-186 31501521-6 2020 Human clinical surveys also linked the expression of TR4, lncTASR, and AXL to the RCC survival, and results from multiple RCC cell lines revealed that targeting this newly identified TR4-mediated signaling with small molecules, including tretinoin, metformin, or TR4-shRNAs, all led to increase the sunitinib sensitivity to better suppress the RCC progression, and our preclinical study using the in vivo mouse model further proved tretinoin had a better synergistic effect to increase sunitinib sensitivity to suppress RCC progression. Sunitinib 486-495 nuclear receptor subfamily 2 group C member 2 Homo sapiens 183-186 31501521-6 2020 Human clinical surveys also linked the expression of TR4, lncTASR, and AXL to the RCC survival, and results from multiple RCC cell lines revealed that targeting this newly identified TR4-mediated signaling with small molecules, including tretinoin, metformin, or TR4-shRNAs, all led to increase the sunitinib sensitivity to better suppress the RCC progression, and our preclinical study using the in vivo mouse model further proved tretinoin had a better synergistic effect to increase sunitinib sensitivity to suppress RCC progression. Sunitinib 486-495 nuclear receptor subfamily 2 group C member 2 Homo sapiens 183-186 31231785-1 2019 Purpose Sunitinib is a vascular endothelial growth factor receptor (VEGFR) inhibitor with antitumor activity against bladder cancer. Sunitinib 8-17 kinase insert domain receptor Homo sapiens 23-66 31231785-1 2019 Purpose Sunitinib is a vascular endothelial growth factor receptor (VEGFR) inhibitor with antitumor activity against bladder cancer. Sunitinib 8-17 kinase insert domain receptor Homo sapiens 68-73 31775817-9 2019 Finally, we show that the FDA-approved protein kinase inhibitor, sunitinib, which has activity against DLK and LZK, is able to produce similar increases in RGC survival. Sunitinib 65-74 mitogen-activated protein kinase kinase kinase 12 Mus musculus 103-106 31775817-9 2019 Finally, we show that the FDA-approved protein kinase inhibitor, sunitinib, which has activity against DLK and LZK, is able to produce similar increases in RGC survival. Sunitinib 65-74 mitogen-activated protein kinase kinase kinase 13 Mus musculus 111-114 31427001-0 2019 HP-1 inhibits the progression of ccRCC and enhances sunitinib therapeutic effects by suppressing EMT. Sunitinib 52-61 haptoglobin Mus musculus 0-4 31427001-7 2019 In addition, the HP-1/sunitinib combination inhibited the PI3K/Akt/VEGFR pathway, reducing the expression of pathway-related proteins. Sunitinib 22-31 haptoglobin Mus musculus 17-21 31427001-7 2019 In addition, the HP-1/sunitinib combination inhibited the PI3K/Akt/VEGFR pathway, reducing the expression of pathway-related proteins. Sunitinib 22-31 thymoma viral proto-oncogene 1 Mus musculus 63-66 31427001-8 2019 The HP-1-induced enhancement of sunitinib effects on tumor growth were also observed in vivo in a xenograft mouse model. Sunitinib 32-41 haptoglobin Mus musculus 4-8 31427001-9 2019 Overall, these results indicated that HP-1 exerted antitumor effects against clear cell renal cell carcinoma (ccRCC) and enhanced the therapeutic efficacy of sunitinib. Sunitinib 158-167 haptoglobin Mus musculus 38-42 31348025-7 2019 Based on the S-TRAC data, sunitinib is approved for use in the adjuvant setting, and can be considered under select circumstances. Sunitinib 26-35 T cell receptor alpha constant Homo sapiens 15-19 31545458-1 2019 The present study aimed to investigate the potential mechanisms of human miR-942 in the sunitinib-resistance of renal cell carcinoma (RCC). Sunitinib 88-97 microRNA 942 Homo sapiens 73-80 31545458-11 2019 The expression level of miR-942 was significantly increased in sunitinib-resistant cells. Sunitinib 63-72 microRNA 942 Homo sapiens 24-31 31471309-6 2019 RESULTS: VEGF and PlGF increased after 4 weeks on sunitinib or sorafenib (P < 0.0001 for both) and returned to baseline at 6 weeks on sunitinib (corresponding to the break in the sunitinib schedule) but not sorafenib (which was administered continuously). Sunitinib 50-59 vascular endothelial growth factor A Homo sapiens 9-13 31471309-10 2019 CXCL10 was elevated at 4 and 6 weeks on sunitinib and sorafenib but not on placebo. Sunitinib 40-49 C-X-C motif chemokine ligand 10 Homo sapiens 0-6 31371341-4 2019 Our primary aim was to correlate progression-free survival (PFS) by independent central review with PD-L1 status in patients treated with cabozantinib, everolimus (METEOR), or sunitinib (CABOSUN). Sunitinib 176-185 CD274 molecule Homo sapiens 100-105 31385548-7 2019 PDGF-BB promoted the proliferation, migration and invasion of hPFs, which can be significantly suppressed by sunitinib via inhibition of the PDGFR-beta/extracellular signal-regulated kinase (ERK) pathway. Sunitinib 109-118 platelet derived growth factor receptor alpha Homo sapiens 141-151 31400051-13 2019 Hypermethylated PON1 promoted migration, invasion and proliferation of sunitinib-resistance renal cancer cells and arrested more cells in G0/G1 phase. Sunitinib 71-80 paraoxonase 1 Homo sapiens 16-20 31445075-8 2019 Accordingly, sunitinib caused caspase 3 activation and DNA fragmentation in H9c2 cells. Sunitinib 13-22 caspase 3 Rattus norvegicus 30-39 31445075-9 2019 Co-exposure with mito-TEMPO (mitochondrial-specific ROS scavenger) for 24 h prevented ATP and GSH depletion, as well as the increases in H2O2 and caspase 3/7 activity observed with sunitinib. Sunitinib 181-190 caspase 3 Rattus norvegicus 146-155 31519573-0 2019 Inhibition of Tumor Growth and Sensitization to Sunitinib by RNA Interference Targeting Programmed Death-ligand 1 in Mouse Renal Cell Carcinoma RenCa Model. Sunitinib 48-57 CD274 antigen Mus musculus 88-113 31519573-2 2019 The purpose of this study was to address the inhibitory effects of programmed death-ligand 1 (PD-L1) expression on growth and sensitivity to sunitinib in the mouse RCC RenCa model. Sunitinib 141-150 CD274 antigen Mus musculus 67-92 31519573-2 2019 The purpose of this study was to address the inhibitory effects of programmed death-ligand 1 (PD-L1) expression on growth and sensitivity to sunitinib in the mouse RCC RenCa model. Sunitinib 141-150 CD274 antigen Mus musculus 94-99 31519573-7 2019 Treatment of mice bearing each tumor with sunitinib resulted in a significant reduction of the RenCa/sh-PD-L1 tumor compared to the RenCa/Co tumor. Sunitinib 42-51 CD274 antigen Mus musculus 104-109 31519573-9 2019 CONCLUSION: Suppressed expression of PD-L1 could increase tumor-infiltrating CD8+ T cells and result in growth inhibition as well as enhanced sensitivity to sunitinib in the RenCa model. Sunitinib 157-166 CD274 antigen Mus musculus 37-42 31467229-0 2019 MiR-376b-3p Is Associated With Long-term Response to Sunitinib in Metastatic Renal Cell Carcinoma Patients. Sunitinib 53-62 microRNA 376b Homo sapiens 0-8 31385548-7 2019 PDGF-BB promoted the proliferation, migration and invasion of hPFs, which can be significantly suppressed by sunitinib via inhibition of the PDGFR-beta/extracellular signal-regulated kinase (ERK) pathway. Sunitinib 109-118 mitogen-activated protein kinase 1 Homo sapiens 191-194 31385548-8 2019 In the ex vivo model, the knockout of PDGFRB and sunitinib treatment blocked the proliferation and motility of fibroblasts in the pterygial stroma via the suppression of PDGFR-beta/ERK pathway. Sunitinib 49-58 platelet derived growth factor receptor alpha Homo sapiens 170-180 31385548-8 2019 In the ex vivo model, the knockout of PDGFRB and sunitinib treatment blocked the proliferation and motility of fibroblasts in the pterygial stroma via the suppression of PDGFR-beta/ERK pathway. Sunitinib 49-58 mitogen-activated protein kinase 1 Homo sapiens 181-184 31385548-9 2019 Conclusion: This study demonstrates that PDGFR-beta may be a potential therapeutic target for pterygium, and inhibition of PDGFR-beta by sunitinib is a promising and effective approach for pterygium treatment. Sunitinib 137-146 platelet derived growth factor receptor alpha Homo sapiens 123-133 31464635-10 2019 The sunitinib-axitinib sequence was safe and effective, the mOS was 41.15 months. Sunitinib 4-13 Moloney sarcoma oncogene Mus musculus 60-63 31401413-9 2019 In vitro, we found that adiponectin inhibited migration, invasion and sensitized RCC cells to sunitinib though interacting with AdipoR1, but not AdipoR2. Sunitinib 94-103 adiponectin, C1Q and collagen domain containing Homo sapiens 24-35 31401413-10 2019 Furthermore, we demonstrated that adiponentin-AdipoR1 axis inhibits tumor cells migration and invasion by blocking the GSK3beta/beta-Catenin pathway and enhances sunitinib sensitivity via abrogating PI3K/AKT/NF-kappaB signaling. Sunitinib 162-171 adiponectin receptor 1 Homo sapiens 46-53 31443471-5 2019 Currently vascular endothelial growth factor (VEGF) targeted therapy based on tyrosine kinase inhibitors (TKI), sunitinib and pazopanib is the alternative regimen for patients who cannot tolerate immune checkpoint inhibitors (ICI). Sunitinib 112-121 vascular endothelial growth factor A Homo sapiens 10-44 31507414-0 2019 Downregulation of miR-146a Contributes to Cardiac Dysfunction Induced by the Tyrosine Kinase Inhibitor Sunitinib. Sunitinib 103-112 microRNA 146 Mus musculus 18-26 31507414-4 2019 A significant downregulation of miR-146a was observed in the myocardium of sunitinib-treated mice, along with a 20% decrease in left ventricle ejection fraction (LVEF). Sunitinib 75-84 microRNA 146 Mus musculus 32-40 31507414-8 2019 Significant upregulation of PLN and ANK2 at the mRNA and protein levels was observed in the myocardium of sunitinib-treated mice. Sunitinib 106-115 phospholamban Mus musculus 28-31 31507414-8 2019 Significant upregulation of PLN and ANK2 at the mRNA and protein levels was observed in the myocardium of sunitinib-treated mice. Sunitinib 106-115 ankyrin 2, brain Mus musculus 36-40 31507414-10 2019 SiRNA knockdown of PLN or ANK2 prevented sunitinib-induced suppression of contractility in hiPSC-CMs. Sunitinib 41-50 phospholamban Mus musculus 19-22 31507414-10 2019 SiRNA knockdown of PLN or ANK2 prevented sunitinib-induced suppression of contractility in hiPSC-CMs. Sunitinib 41-50 ankyrin 2, brain Mus musculus 26-30 31507414-11 2019 Therefore, our in vivo and in vitro results showed that sunitinib downregulated miR-146a, which contributes to cardiac contractile dysfunction by regulating the downstream targets PLN and ANK2, and that upregulation of miR-146a alleviated the inhibitory effect of SNT on cardiac contractility. Sunitinib 56-65 microRNA 146 Mus musculus 80-88 31507414-11 2019 Therefore, our in vivo and in vitro results showed that sunitinib downregulated miR-146a, which contributes to cardiac contractile dysfunction by regulating the downstream targets PLN and ANK2, and that upregulation of miR-146a alleviated the inhibitory effect of SNT on cardiac contractility. Sunitinib 56-65 phospholamban Mus musculus 180-183 31507414-11 2019 Therefore, our in vivo and in vitro results showed that sunitinib downregulated miR-146a, which contributes to cardiac contractile dysfunction by regulating the downstream targets PLN and ANK2, and that upregulation of miR-146a alleviated the inhibitory effect of SNT on cardiac contractility. Sunitinib 56-65 ankyrin 2, brain Mus musculus 188-192 31507414-11 2019 Therefore, our in vivo and in vitro results showed that sunitinib downregulated miR-146a, which contributes to cardiac contractile dysfunction by regulating the downstream targets PLN and ANK2, and that upregulation of miR-146a alleviated the inhibitory effect of SNT on cardiac contractility. Sunitinib 56-65 microRNA 146 Mus musculus 219-227 31462260-11 2019 Mechanistic studies confirmed that HM-3 and Sunitinib regulated distribution of integrin alphavbeta3, alpha5beta1, VEGFR2 and alphavbeta3-VEGFR2 complexes, both inside and outside of the lipid raft regions to regulate endothelial cell migration and intracellular RhoGTPase activities. Sunitinib 44-53 kinase insert domain protein receptor Mus musculus 115-121 31462260-11 2019 Mechanistic studies confirmed that HM-3 and Sunitinib regulated distribution of integrin alphavbeta3, alpha5beta1, VEGFR2 and alphavbeta3-VEGFR2 complexes, both inside and outside of the lipid raft regions to regulate endothelial cell migration and intracellular RhoGTPase activities. Sunitinib 44-53 kinase insert domain protein receptor Mus musculus 138-144 31507414-11 2019 Therefore, our in vivo and in vitro results showed that sunitinib downregulated miR-146a, which contributes to cardiac contractile dysfunction by regulating the downstream targets PLN and ANK2, and that upregulation of miR-146a alleviated the inhibitory effect of SNT on cardiac contractility. Sunitinib 56-65 fibroblast growth factor receptor substrate 2 Homo sapiens 264-267 31507414-12 2019 Thus, miR-146a could be a useful protective agent against sunitinib-induced cardiac dysfunction. Sunitinib 58-67 microRNA 146 Mus musculus 6-14 31443471-5 2019 Currently vascular endothelial growth factor (VEGF) targeted therapy based on tyrosine kinase inhibitors (TKI), sunitinib and pazopanib is the alternative regimen for patients who cannot tolerate immune checkpoint inhibitors (ICI). Sunitinib 112-121 vascular endothelial growth factor A Homo sapiens 46-50 31411627-3 2019 The pH-sensitive micelle core encapsulates PTX, while beta-cyclodextrin molecules being conjugated to the micelle shell via matrix metalloproteinase 2 (MMP-2) sensitive peptides include sunitinib. Sunitinib 186-195 matrix metallopeptidase 2 Homo sapiens 152-157 31411627-4 2019 Following the pH and MMP-2 dual sensitive structure design, the micelle may sequentially release sunitinib inside the tumor extracellular matrix and PTX into cancer cells through responding to enriched MMP-2 levels and decreased pH, respectively. Sunitinib 97-106 matrix metallopeptidase 2 Homo sapiens 21-26 31411627-4 2019 Following the pH and MMP-2 dual sensitive structure design, the micelle may sequentially release sunitinib inside the tumor extracellular matrix and PTX into cancer cells through responding to enriched MMP-2 levels and decreased pH, respectively. Sunitinib 97-106 matrix metallopeptidase 2 Homo sapiens 202-207 31294623-9 2019 Sunitinib therapy substantially mitigated both AAA formation and further progression of established AAAs, attenuated aneurysmal aortic MMP2 (matrix metalloproteinase) and MMP9 protein expression, inhibited inflammatory monocyte and neutrophil chemotaxis to VEGF-A, and reduced MMP2, MMP9, and VEGF-A mRNA expression in macrophages and smooth muscle cells in vitro. Sunitinib 0-9 matrix metallopeptidase 2 Mus musculus 135-139 31534544-0 2019 DNA methylation-regulated QPCT promotes sunitinib resistance by increasing HRAS stability in renal cell carcinoma. Sunitinib 40-49 glutaminyl-peptide cyclotransferase Homo sapiens 26-30 31534544-0 2019 DNA methylation-regulated QPCT promotes sunitinib resistance by increasing HRAS stability in renal cell carcinoma. Sunitinib 40-49 HRas proto-oncogene, GTPase Homo sapiens 75-79 31534544-4 2019 We verified glutaminyl peptide cyclotransferase (QPCT) expression in sunitinib-nonresponsive and -responsive RCC tissues via qRT-PCR, western blot and immunohistochemical assays. Sunitinib 69-78 glutaminyl-peptide cyclotransferase Homo sapiens 12-47 31534544-4 2019 We verified glutaminyl peptide cyclotransferase (QPCT) expression in sunitinib-nonresponsive and -responsive RCC tissues via qRT-PCR, western blot and immunohistochemical assays. Sunitinib 69-78 glutaminyl-peptide cyclotransferase Homo sapiens 49-53 31534544-5 2019 Then, cell counting kit 8 (CCK-8), plate colony formation and flow cytometric assays were used to verify the function of QPCT in RCC sunitinib resistance after QPCT intervention or overexpression. Sunitinib 133-142 glutaminyl-peptide cyclotransferase Homo sapiens 121-125 31534544-8 2019 Results: We found that the degree of methylation in the QPCT promoter region was significantly different between sunitinib-nonresponsive and -responsive RCC tissues. Sunitinib 113-122 glutaminyl-peptide cyclotransferase Homo sapiens 56-60 31534544-9 2019 In the sunitinib-nonresponsive tissues, the degree of methylation in the QPCT promoter region was significantly reduced, and the expression of QPCT was upregulated, which correlated with a clinically poor response to sunitinib. Sunitinib 7-16 glutaminyl-peptide cyclotransferase Homo sapiens 73-77 31534544-9 2019 In the sunitinib-nonresponsive tissues, the degree of methylation in the QPCT promoter region was significantly reduced, and the expression of QPCT was upregulated, which correlated with a clinically poor response to sunitinib. Sunitinib 7-16 glutaminyl-peptide cyclotransferase Homo sapiens 143-147 31534544-9 2019 In the sunitinib-nonresponsive tissues, the degree of methylation in the QPCT promoter region was significantly reduced, and the expression of QPCT was upregulated, which correlated with a clinically poor response to sunitinib. Sunitinib 217-226 glutaminyl-peptide cyclotransferase Homo sapiens 143-147 31534544-10 2019 A knockdown of QPCT conferred sunitinib sensitivity traits to RCC cells, whereas an overexpression of QPCT restored sunitinib resistance in RCC cells. Sunitinib 116-125 glutaminyl-peptide cyclotransferase Homo sapiens 102-106 31534544-13 2019 Conclusion: QPCT may be a novel predictor of the response to sunitinib therapy in RCC patients and a potential therapeutic target. Sunitinib 61-70 glutaminyl-peptide cyclotransferase Homo sapiens 12-16 31294623-9 2019 Sunitinib therapy substantially mitigated both AAA formation and further progression of established AAAs, attenuated aneurysmal aortic MMP2 (matrix metalloproteinase) and MMP9 protein expression, inhibited inflammatory monocyte and neutrophil chemotaxis to VEGF-A, and reduced MMP2, MMP9, and VEGF-A mRNA expression in macrophages and smooth muscle cells in vitro. Sunitinib 0-9 matrix metallopeptidase 9 Mus musculus 171-175 31294623-9 2019 Sunitinib therapy substantially mitigated both AAA formation and further progression of established AAAs, attenuated aneurysmal aortic MMP2 (matrix metalloproteinase) and MMP9 protein expression, inhibited inflammatory monocyte and neutrophil chemotaxis to VEGF-A, and reduced MMP2, MMP9, and VEGF-A mRNA expression in macrophages and smooth muscle cells in vitro. Sunitinib 0-9 vascular endothelial growth factor A Mus musculus 257-263 31294623-9 2019 Sunitinib therapy substantially mitigated both AAA formation and further progression of established AAAs, attenuated aneurysmal aortic MMP2 (matrix metalloproteinase) and MMP9 protein expression, inhibited inflammatory monocyte and neutrophil chemotaxis to VEGF-A, and reduced MMP2, MMP9, and VEGF-A mRNA expression in macrophages and smooth muscle cells in vitro. Sunitinib 0-9 matrix metallopeptidase 2 Mus musculus 277-281 31294623-9 2019 Sunitinib therapy substantially mitigated both AAA formation and further progression of established AAAs, attenuated aneurysmal aortic MMP2 (matrix metalloproteinase) and MMP9 protein expression, inhibited inflammatory monocyte and neutrophil chemotaxis to VEGF-A, and reduced MMP2, MMP9, and VEGF-A mRNA expression in macrophages and smooth muscle cells in vitro. Sunitinib 0-9 matrix metallopeptidase 9 Mus musculus 283-287 31294623-9 2019 Sunitinib therapy substantially mitigated both AAA formation and further progression of established AAAs, attenuated aneurysmal aortic MMP2 (matrix metalloproteinase) and MMP9 protein expression, inhibited inflammatory monocyte and neutrophil chemotaxis to VEGF-A, and reduced MMP2, MMP9, and VEGF-A mRNA expression in macrophages and smooth muscle cells in vitro. Sunitinib 0-9 vascular endothelial growth factor A Mus musculus 293-299 30644033-0 2019 Carnitine Supplementation Attenuates Sunitinib-Induced Inhibition of AMP-Activated Protein Kinase Downstream Signals in Cardiac Tissues. Sunitinib 37-46 protein kinase AMP-activated catalytic subunit alpha 2 Rattus norvegicus 69-97 31423209-0 2019 Significance of cyclooxygenase-2, prostaglandin E2 and CD133 levels in sunitinib-resistant renal cell carcinoma. Sunitinib 71-80 prostaglandin-endoperoxide synthase 2 Homo sapiens 16-32 31268155-2 2019 The Von Hippel-Lindau tumor suppressor (VHL)-hypoxia-inducible factor 1 subunit alpha (HIF1A)/hypoxia-inducible factor 2alpha (HIF2A)-vascular endothelial growth factor A (VEGFA) protein axis is involved in the development and progression of ccRCC, whereas sunitinib, a tyrosine kinase inhibitor, blocks the binding of VEGFA to its receptor. Sunitinib 257-266 von Hippel-Lindau tumor suppressor Homo sapiens 40-43 31268155-2 2019 The Von Hippel-Lindau tumor suppressor (VHL)-hypoxia-inducible factor 1 subunit alpha (HIF1A)/hypoxia-inducible factor 2alpha (HIF2A)-vascular endothelial growth factor A (VEGFA) protein axis is involved in the development and progression of ccRCC, whereas sunitinib, a tyrosine kinase inhibitor, blocks the binding of VEGFA to its receptor. Sunitinib 257-266 vascular endothelial growth factor A Homo sapiens 134-170 31268155-3 2019 The aim of the present study was to examine the possible association of the gene expression of VHL, HIF1A, HIF2A, VEGFA and tumor protein P53 (P53) in cancer tissue with the outcome of ccRCC patients who were treated with sunitinib as first-line therapy following nephrectomy. Sunitinib 222-231 von Hippel-Lindau tumor suppressor Homo sapiens 95-98 31268155-3 2019 The aim of the present study was to examine the possible association of the gene expression of VHL, HIF1A, HIF2A, VEGFA and tumor protein P53 (P53) in cancer tissue with the outcome of ccRCC patients who were treated with sunitinib as first-line therapy following nephrectomy. Sunitinib 222-231 tumor protein p53 Homo sapiens 138-141 31268155-3 2019 The aim of the present study was to examine the possible association of the gene expression of VHL, HIF1A, HIF2A, VEGFA and tumor protein P53 (P53) in cancer tissue with the outcome of ccRCC patients who were treated with sunitinib as first-line therapy following nephrectomy. Sunitinib 222-231 tumor protein p53 Homo sapiens 143-146 31268155-8 2019 Sunitinib resistance was associated with high HIF2A and VEGFA protein levels (b=0.57 and b=0.69 for OS and PFS, respectively; P<0.001). Sunitinib 0-9 endothelial PAS domain protein 1 Homo sapiens 46-51 31268155-8 2019 Sunitinib resistance was associated with high HIF2A and VEGFA protein levels (b=0.57 and b=0.69 for OS and PFS, respectively; P<0.001). Sunitinib 0-9 vascular endothelial growth factor A Homo sapiens 56-61 31339065-3 2019 This Review focuses on the existing evidence on patient-reported outcomes and health-related quality of life with several tyrosine kinase inhibitors (pazopanib, sunitinib, cabozantinib and tivozanib) used as first-line treatment for metastatic renal cell carcinoma. Sunitinib 161-170 TXK tyrosine kinase Homo sapiens 122-137 31392045-15 2019 Nine patients received sunitinib, of them 3 had germline mutation (2 in MEN1 and 1 in VHL genes). Sunitinib 23-32 menin 1 Homo sapiens 72-82 31423209-0 2019 Significance of cyclooxygenase-2, prostaglandin E2 and CD133 levels in sunitinib-resistant renal cell carcinoma. Sunitinib 71-80 prominin 1 Homo sapiens 55-60 31423209-13 2019 In summary, the results indicate that activation of the COX-2-PGE2 pathway in RCC leads to the development of sunitinib resistance and may serve an important role in the maintenance of the characteristics of stem cells that are closely associated with drug resistance. Sunitinib 110-119 mitochondrially encoded cytochrome c oxidase II Homo sapiens 56-66 31366950-0 2019 SQSTM1/p62 loss reverses the inhibitory effect of sunitinib on autophagy independent of AMPK signaling. Sunitinib 50-59 sequestosome 1 Homo sapiens 0-6 31366950-0 2019 SQSTM1/p62 loss reverses the inhibitory effect of sunitinib on autophagy independent of AMPK signaling. Sunitinib 50-59 sequestosome 1 Homo sapiens 7-10 30875096-8 2019 Activation of PPARalpha and inhibition of xenobiotic metabolism may be of value in attenuating sunitinib hepatotoxicity. Sunitinib 95-104 peroxisome proliferator activated receptor alpha Mus musculus 14-23 31054432-7 2019 IC50 of the promising compounds were in the range of 247-793 nM for VEGFR-2 in reference to sunitinib (IC50 320 nM), and 369-725 nM for EGFR in reference to erlotinib (IC50 568 nM). Sunitinib 92-101 kinase insert domain receptor Homo sapiens 68-75 31054432-7 2019 IC50 of the promising compounds were in the range of 247-793 nM for VEGFR-2 in reference to sunitinib (IC50 320 nM), and 369-725 nM for EGFR in reference to erlotinib (IC50 568 nM). Sunitinib 92-101 epidermal growth factor receptor Homo sapiens 69-73 31319613-5 2019 The administration of a small molecule inhibitor of CAIX, SLC-0111, significantly reduced overall metastatic burden, whereas exposure to sunitinib increased hypoxia and CAIX expression in primary tumors, and failed to inhibit metastasis. Sunitinib 137-146 carbonic anhydrase 9 Homo sapiens 169-173 31319613-7 2019 Furthermore, combining sunitinib and SLC-0111 significantly reduced both primary tumor growth and sunitinib-induced metastasis to the lung. Sunitinib 98-107 C-C motif chemokine ligand 21 Homo sapiens 37-40 31039345-2 2019 A tyrosine kinase inhibitor (TKIs), sunitinib, was screened in our group previously and showed antagonistic activity for cytokine release in a TLR7 stimulation model. Sunitinib 36-45 toll-like receptor 7 Mus musculus 143-147 31039345-4 2019 In vitro, nearly all of the eleven TKIs decreased the TNF-alpha levels induced by the TLR7 agonist, especially sunitinib. Sunitinib 111-120 tumor necrosis factor Mus musculus 54-63 31039345-4 2019 In vitro, nearly all of the eleven TKIs decreased the TNF-alpha levels induced by the TLR7 agonist, especially sunitinib. Sunitinib 111-120 toll-like receptor 7 Mus musculus 86-90 31039345-5 2019 Furthermore, sunitinib displayed potent inhibition of the cytokine levels triggered by several types of TLR ligands, including TLR3, TLR4, TLR7/8 and TLR9, in mouse spleen lymphocytes, mouse BMDCs and human PBMCs. Sunitinib 13-22 toll-like receptor 3 Mus musculus 127-131 31039345-5 2019 Furthermore, sunitinib displayed potent inhibition of the cytokine levels triggered by several types of TLR ligands, including TLR3, TLR4, TLR7/8 and TLR9, in mouse spleen lymphocytes, mouse BMDCs and human PBMCs. Sunitinib 13-22 toll-like receptor 4 Mus musculus 133-137 31039345-5 2019 Furthermore, sunitinib displayed potent inhibition of the cytokine levels triggered by several types of TLR ligands, including TLR3, TLR4, TLR7/8 and TLR9, in mouse spleen lymphocytes, mouse BMDCs and human PBMCs. Sunitinib 13-22 toll-like receptor 7 Mus musculus 139-145 31039345-5 2019 Furthermore, sunitinib displayed potent inhibition of the cytokine levels triggered by several types of TLR ligands, including TLR3, TLR4, TLR7/8 and TLR9, in mouse spleen lymphocytes, mouse BMDCs and human PBMCs. Sunitinib 13-22 toll-like receptor 9 Mus musculus 150-154 31039345-6 2019 The in vivo results showed that sunitinib efficiently depressed the LPS-induced cytokine storm, i.e., rapid and intense production of TNF-alpha and IL-6. Sunitinib 32-41 tumor necrosis factor Mus musculus 134-143 31039345-6 2019 The in vivo results showed that sunitinib efficiently depressed the LPS-induced cytokine storm, i.e., rapid and intense production of TNF-alpha and IL-6. Sunitinib 32-41 interleukin 6 Mus musculus 148-152 31039345-8 2019 As for the immunosuppressive mechanisms of sunitinib, at least the PDGFR-activated ERK and p38 pathways were critical, although we could not rule out the possibility of other pathways being involved. Sunitinib 43-52 platelet derived growth factor receptor, beta polypeptide Mus musculus 67-72 31039345-8 2019 As for the immunosuppressive mechanisms of sunitinib, at least the PDGFR-activated ERK and p38 pathways were critical, although we could not rule out the possibility of other pathways being involved. Sunitinib 43-52 Eph receptor B2 Mus musculus 83-86 31039345-8 2019 As for the immunosuppressive mechanisms of sunitinib, at least the PDGFR-activated ERK and p38 pathways were critical, although we could not rule out the possibility of other pathways being involved. Sunitinib 43-52 mitogen-activated protein kinase 14 Mus musculus 91-94 31297023-6 2019 Moreover, a molecular docking simulation was performed for compound 16 and sunitinib to predict the protein-ligand interactions with the active site of VEGFR2. Sunitinib 75-84 kinase insert domain receptor Homo sapiens 152-158 30914797-5 2019 Only two trials of an autologous renal tumour cell vaccine and of the vascular endothelial growth factor receptor (VEGFR) tyrosine kinase inhibitor sunitinib have shown positive results, but these have been criticized for methodological reasons and conflicting data, respectively. Sunitinib 148-157 kinase insert domain receptor Homo sapiens 70-113 30914797-5 2019 Only two trials of an autologous renal tumour cell vaccine and of the vascular endothelial growth factor receptor (VEGFR) tyrosine kinase inhibitor sunitinib have shown positive results, but these have been criticized for methodological reasons and conflicting data, respectively. Sunitinib 148-157 kinase insert domain receptor Homo sapiens 115-120 30806670-6 2019 Sunitinib exerts its regenerative effects via transient inhibition of SHP-2 and subsequent activation of the STAT3 pathway. Sunitinib 0-9 protein tyrosine phosphatase, non-receptor type 11 Mus musculus 70-75 30806670-6 2019 Sunitinib exerts its regenerative effects via transient inhibition of SHP-2 and subsequent activation of the STAT3 pathway. Sunitinib 0-9 signal transducer and activator of transcription 3 Mus musculus 109-114 31018963-0 2019 Multiple conformational states of the HPK1 kinase domain in complex with sunitinib reveal the structural changes accompanying HPK1 trans-regulation. Sunitinib 73-82 mitogen-activated protein kinase kinase kinase kinase 1 Homo sapiens 38-42 31293642-0 2019 Overexpression of miR-15b Promotes Resistance to Sunitinib in Renal Cell Carcinoma. Sunitinib 49-58 microRNA 15b Mus musculus 18-25 31293642-5 2019 Results: Reproduction of GEO datasets revealed miR-15b significantly upregulated in sunitinib- resistant ccRCC. Sunitinib 84-93 microRNA 15b Mus musculus 47-54 31293642-6 2019 Five out of seven ccRCC cell lines demonstrated significantly overexpressed miR-15b after sunitinib treatment. Sunitinib 90-99 microRNA 15b Mus musculus 76-83 31293642-7 2019 Vector-mediated overexpression of miR-15b significantly induced resistance to sunitinib in ccRCC cells. Sunitinib 78-87 microRNA 15b Mus musculus 34-41 31293642-8 2019 Overexpression of miR-15b significantly induced less population in G1 phase of cell cycle and less apoptosis in cells treated sunitinib. Sunitinib 126-135 microRNA 15b Mus musculus 18-25 31293642-12 2019 Silencing of CCNC mimicked overexpression of miR-25 inducing cell cycle progression passing G1 phase and less apoptosis in ccRCC cells treated by sunitinib. Sunitinib 146-155 cyclin C Mus musculus 13-17 31293642-12 2019 Silencing of CCNC mimicked overexpression of miR-25 inducing cell cycle progression passing G1 phase and less apoptosis in ccRCC cells treated by sunitinib. Sunitinib 146-155 microRNA 25 Mus musculus 45-51 31248045-0 2019 Heightened JNK Activation and Reduced XIAP Levels Promote TRAIL and Sunitinib-Mediated Apoptosis in Colon Cancer Models. Sunitinib 68-77 X-linked inhibitor of apoptosis Homo sapiens 38-42 31248045-3 2019 We hypothesized that a disruption of pro-survival signaling cascades with the multi-tyrosine kinase inhibitor sunitinib and would be an effective strategy to enhance TRAIL-mediated apoptosis. Sunitinib 110-119 TNF superfamily member 10 Homo sapiens 166-171 31248045-4 2019 Here we demonstrate that sunitinib significantly augments the anticancer activity of TRAIL in models of colon cancer. Sunitinib 25-34 TNF superfamily member 10 Homo sapiens 85-90 31248045-5 2019 The therapeutic benefit of the TRAIL/sunitinib combination was associated with increased apoptosis marked by enhanced caspase-3 cleavage and DNA fragmentation. Sunitinib 37-46 caspase 3 Homo sapiens 118-127 31248045-6 2019 Overexpression of the anti-apoptotic factor B-cell lymphoma 2 (BCL-2) in HCT116 cells reduced TRAIL/sunitinib-mediated apoptosis, further supporting that sunitinib enhances the anticancer activity of TRAIL via augmented apoptosis. Sunitinib 100-109 BCL2 apoptosis regulator Homo sapiens 44-61 31248045-6 2019 Overexpression of the anti-apoptotic factor B-cell lymphoma 2 (BCL-2) in HCT116 cells reduced TRAIL/sunitinib-mediated apoptosis, further supporting that sunitinib enhances the anticancer activity of TRAIL via augmented apoptosis. Sunitinib 100-109 BCL2 apoptosis regulator Homo sapiens 63-68 31248045-6 2019 Overexpression of the anti-apoptotic factor B-cell lymphoma 2 (BCL-2) in HCT116 cells reduced TRAIL/sunitinib-mediated apoptosis, further supporting that sunitinib enhances the anticancer activity of TRAIL via augmented apoptosis. Sunitinib 154-163 BCL2 apoptosis regulator Homo sapiens 44-61 31248045-6 2019 Overexpression of the anti-apoptotic factor B-cell lymphoma 2 (BCL-2) in HCT116 cells reduced TRAIL/sunitinib-mediated apoptosis, further supporting that sunitinib enhances the anticancer activity of TRAIL via augmented apoptosis. Sunitinib 154-163 BCL2 apoptosis regulator Homo sapiens 63-68 31248045-6 2019 Overexpression of the anti-apoptotic factor B-cell lymphoma 2 (BCL-2) in HCT116 cells reduced TRAIL/sunitinib-mediated apoptosis, further supporting that sunitinib enhances the anticancer activity of TRAIL via augmented apoptosis. Sunitinib 154-163 TNF superfamily member 10 Homo sapiens 94-99 31248045-6 2019 Overexpression of the anti-apoptotic factor B-cell lymphoma 2 (BCL-2) in HCT116 cells reduced TRAIL/sunitinib-mediated apoptosis, further supporting that sunitinib enhances the anticancer activity of TRAIL via augmented apoptosis. Sunitinib 154-163 TNF superfamily member 10 Homo sapiens 200-205 31248045-7 2019 Analysis of pro-survival factors identified that the combination of TRAIL and sunitinib significantly downregulated the anti-apoptotic protein X-linked inhibitor of apoptosis protein (XIAP) through a c-Jun N-terminal kinase (JNK)-mediated mechanism. Sunitinib 78-87 X-linked inhibitor of apoptosis Homo sapiens 143-182 31248045-7 2019 Analysis of pro-survival factors identified that the combination of TRAIL and sunitinib significantly downregulated the anti-apoptotic protein X-linked inhibitor of apoptosis protein (XIAP) through a c-Jun N-terminal kinase (JNK)-mediated mechanism. Sunitinib 78-87 X-linked inhibitor of apoptosis Homo sapiens 184-188 31248045-7 2019 Analysis of pro-survival factors identified that the combination of TRAIL and sunitinib significantly downregulated the anti-apoptotic protein X-linked inhibitor of apoptosis protein (XIAP) through a c-Jun N-terminal kinase (JNK)-mediated mechanism. Sunitinib 78-87 mitogen-activated protein kinase 8 Homo sapiens 200-223 31248045-7 2019 Analysis of pro-survival factors identified that the combination of TRAIL and sunitinib significantly downregulated the anti-apoptotic protein X-linked inhibitor of apoptosis protein (XIAP) through a c-Jun N-terminal kinase (JNK)-mediated mechanism. Sunitinib 78-87 mitogen-activated protein kinase 8 Homo sapiens 225-228 31248045-8 2019 Short hairpin RNA (shRNA)-mediated knockdown of JNK confirmed its key role in the regulation of sensitivity to this combination as cells with suppressed JNK expression exhibited significantly reduced TRAIL/sunitinib-mediated apoptosis. Sunitinib 206-215 mitogen-activated protein kinase 8 Homo sapiens 48-51 31248045-8 2019 Short hairpin RNA (shRNA)-mediated knockdown of JNK confirmed its key role in the regulation of sensitivity to this combination as cells with suppressed JNK expression exhibited significantly reduced TRAIL/sunitinib-mediated apoptosis. Sunitinib 206-215 mitogen-activated protein kinase 8 Homo sapiens 153-156 31248045-10 2019 Collectively, our data demonstrate that sunitinib enhances TRAIL-mediated apoptosis by heightened JNK activation, diminished XIAP levels, and augmented apoptosis. Sunitinib 40-49 TNF superfamily member 10 Homo sapiens 59-64 31248045-10 2019 Collectively, our data demonstrate that sunitinib enhances TRAIL-mediated apoptosis by heightened JNK activation, diminished XIAP levels, and augmented apoptosis. Sunitinib 40-49 mitogen-activated protein kinase 8 Homo sapiens 98-101 31248045-10 2019 Collectively, our data demonstrate that sunitinib enhances TRAIL-mediated apoptosis by heightened JNK activation, diminished XIAP levels, and augmented apoptosis. Sunitinib 40-49 X-linked inhibitor of apoptosis Homo sapiens 125-129 31293642-13 2019 Overexpression of miR-15b also counteracted suppression of migration and colony formation by sunitinib in ccRCC cell lines. Sunitinib 93-102 microRNA 15b Mus musculus 18-25 31293642-14 2019 In vivo mouse xenograft models showed recovered tumor growth with miR-15b expression in mice treated with sunitinib. Sunitinib 106-115 microRNA 15b Mus musculus 66-73 31293642-15 2019 Conclusion: We here show miR-15b as a possible culprit for sunitinib resistance in ccRCC. Sunitinib 59-68 microRNA 15b Mus musculus 25-32 31018963-0 2019 Multiple conformational states of the HPK1 kinase domain in complex with sunitinib reveal the structural changes accompanying HPK1 trans-regulation. Sunitinib 73-82 mitogen-activated protein kinase kinase kinase kinase 1 Homo sapiens 126-130 31289593-0 2019 Tumoral Pyruvate Kinase L/R as a Predictive Marker for the Treatment of Renal Cancer Patients with Sunitinib and Sorafenib. Sunitinib 99-108 pyruvate kinase L/R Homo sapiens 8-27 31289593-4 2019 We have previously shown that the tumoral expression of cubilin (CUBN) is associated with progression free survival (PFS) in mRCC patients treated with sunitinib and sorafenib. Sunitinib 152-161 cubilin Homo sapiens 56-63 31289593-4 2019 We have previously shown that the tumoral expression of cubilin (CUBN) is associated with progression free survival (PFS) in mRCC patients treated with sunitinib and sorafenib. Sunitinib 152-161 cubilin Homo sapiens 65-69 29792121-0 2019 ABCG2 Polymorphism rs2231142 and hypothyroidism in metastatic renal cell carcinoma patients treated with sunitinib. Sunitinib 105-114 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 0-5 31289593-14 2019 Conclusions: In this real world study we show that tumoral PKLR membrane expression is a positive predictive biomarker for sunitinib and sorafenib treatment in patients suffering from mRCC. Sunitinib 123-132 pyruvate kinase L/R Homo sapiens 59-63 29792121-10 2019 CONCLUSION: Polymorphism rs2231142 in the efflux pump ABCG2 is associated with hypothyroidism in mRCC patients treated with sunitinib. Sunitinib 124-133 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 54-59 31025080-1 2019 Sunitinib (SNT) is a multi-targeted receptor tyrosine kinase inhibitor that has been approved by the FDA for cancer therapy. Sunitinib 0-9 ret proto-oncogene Homo sapiens 36-60 30707374-6 2019 KIT inhibitors that have been investigated in relevant clinical trials in advanced melanoma include imatinib, sunitinib, dasatinib, and nilotinib. Sunitinib 110-119 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 0-3 31025080-1 2019 Sunitinib (SNT) is a multi-targeted receptor tyrosine kinase inhibitor that has been approved by the FDA for cancer therapy. Sunitinib 11-14 ret proto-oncogene Homo sapiens 36-60 31025080-7 2019 Oral SNT administration (40 mg/kg/day) in mice progressively decreased the PI3K activity and cardiac function in 2 weeks with a significant decrease in the expression and activity of Cav1.2 and SERCA. Sunitinib 5-8 calcium channel, voltage-dependent, L type, alpha 1C subunit Mus musculus 183-189 30837208-0 2019 Metastatic Clear-cell Renal Cell Carcinoma With a Long-term Response to Sunitinib: A Distinct Phenotype Independently Associated With Low PD-L1 Expression. Sunitinib 72-81 CD274 molecule Homo sapiens 138-143 30953637-8 2019 Moreover, the combination of SN-38 and sunitinib caused synergism on colon cancer cells, with significant inhibition of the ABCG2 gene expression and an increase of SN-38 intracellular concentrations. Sunitinib 39-48 ATP binding cassette subfamily G member 2 (Junior blood group) Mus musculus 124-129 31035254-0 2019 EGF and IGF1 affect sunitinib activity in BP-NEN: new putative targets beyond VEGFR? Sunitinib 20-29 epidermal growth factor Homo sapiens 0-3 31035254-0 2019 EGF and IGF1 affect sunitinib activity in BP-NEN: new putative targets beyond VEGFR? Sunitinib 20-29 insulin like growth factor 1 Homo sapiens 8-12 31035254-7 2019 Our results showed that after treatment with sunitinib and/or IGF1, EGF and VEGF, the antiproliferative effect of sunitinib was counteracted by EGF and IGF1 but not by VEGF. Sunitinib 45-54 insulin like growth factor 1 Homo sapiens 152-156 31035254-7 2019 Our results showed that after treatment with sunitinib and/or IGF1, EGF and VEGF, the antiproliferative effect of sunitinib was counteracted by EGF and IGF1 but not by VEGF. Sunitinib 114-123 insulin like growth factor 1 Homo sapiens 62-66 31035254-7 2019 Our results showed that after treatment with sunitinib and/or IGF1, EGF and VEGF, the antiproliferative effect of sunitinib was counteracted by EGF and IGF1 but not by VEGF. Sunitinib 114-123 epidermal growth factor Homo sapiens 68-80 31035254-7 2019 Our results showed that after treatment with sunitinib and/or IGF1, EGF and VEGF, the antiproliferative effect of sunitinib was counteracted by EGF and IGF1 but not by VEGF. Sunitinib 114-123 epidermal growth factor Homo sapiens 68-71 31035254-7 2019 Our results showed that after treatment with sunitinib and/or IGF1, EGF and VEGF, the antiproliferative effect of sunitinib was counteracted by EGF and IGF1 but not by VEGF. Sunitinib 114-123 insulin like growth factor 1 Homo sapiens 152-156 31035254-7 2019 Our results showed that after treatment with sunitinib and/or IGF1, EGF and VEGF, the antiproliferative effect of sunitinib was counteracted by EGF and IGF1 but not by VEGF. Sunitinib 114-123 vascular endothelial growth factor A Homo sapiens 76-80 31035254-8 2019 Therefore, we evaluated with AlphaScreen technology the phosphorylated EGFR and IGF1R levels in primary cultures treated with sunitinib and/or EGF and IGF1. Sunitinib 126-135 epidermal growth factor receptor Homo sapiens 71-75 31035254-8 2019 Therefore, we evaluated with AlphaScreen technology the phosphorylated EGFR and IGF1R levels in primary cultures treated with sunitinib and/or EGF and IGF1. Sunitinib 126-135 insulin like growth factor 1 receptor Homo sapiens 80-85 31035254-8 2019 Therefore, we evaluated with AlphaScreen technology the phosphorylated EGFR and IGF1R levels in primary cultures treated with sunitinib and/or EGF and IGF1. Sunitinib 126-135 epidermal growth factor Homo sapiens 71-74 31035254-8 2019 Therefore, we evaluated with AlphaScreen technology the phosphorylated EGFR and IGF1R levels in primary cultures treated with sunitinib and/or EGF and IGF1. Sunitinib 126-135 insulin like growth factor 1 Homo sapiens 80-84 31035254-9 2019 Results showed a decrease of p-IGF1R after treatment with sunitinib and an increase after co-treatment with IGF1. Sunitinib 58-67 insulin like growth factor 1 receptor Homo sapiens 31-36 31035254-9 2019 Results showed a decrease of p-IGF1R after treatment with sunitinib and an increase after co-treatment with IGF1. Sunitinib 58-67 insulin like growth factor 1 Homo sapiens 31-35 31186733-0 2019 Effect of the Nrf2-ARE signaling pathway on biological characteristics and sensitivity to sunitinib in renal cell carcinoma. Sunitinib 90-99 NFE2 like bZIP transcription factor 2 Homo sapiens 14-18 31186733-7 2019 Following shRNA-Nrf2 transfection, the 786-0 cells demonstrated a significant decrease in viability, cell invasion and scratch healing rate, and the mRNA and protein expression levels of Nrf2, NQO1, HO-1 and glutathione transferase were significantly decreased, which enhanced the sensitivity to sunitinib, arrested cells in the G0/G1 phase and increased apoptosis. Sunitinib 296-305 NFE2 like bZIP transcription factor 2 Homo sapiens 16-20 31217744-5 2019 After exposure to sunitinib (a kind of tyrosine kinase inhibitor, TKI), cell proliferation inhibition was tested by MTT, cell cycle and apoptosis were measured by FCM, autophagy was assessed by fluorescence microscopy and immunoblotting. Sunitinib 18-27 TXK tyrosine kinase Homo sapiens 39-54 31217744-8 2019 After sunitinib inhibited the c-KIT activity, the colony formation efficiency was reduced, and the half-maximal inhibitory concentration (IC50) of sunitinib was low (0.44+-0.17muM) at 48 hours. Sunitinib 6-15 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 30-35 31217744-8 2019 After sunitinib inhibited the c-KIT activity, the colony formation efficiency was reduced, and the half-maximal inhibitory concentration (IC50) of sunitinib was low (0.44+-0.17muM) at 48 hours. Sunitinib 147-156 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 30-35 31054006-3 2019 The S-TRAC study demonstrated a statistically significant improvement in disease-free survival (DFS) of greater than 1 year with adjuvant sunitinib compared to placebo in patients with high-risk localized RCC and earned it FDA approval. Sunitinib 138-147 T cell receptor alpha constant Homo sapiens 6-10 30689282-0 2019 Exploring binding mechanisms of VEGFR2 with three drugs lenvatinib, sorafenib, and sunitinib by molecular dynamics simulation and free energy calculation. Sunitinib 83-92 kinase insert domain receptor Homo sapiens 32-38 30689282-1 2019 Lenvatinib (LEN), sorafenib (SOR), and sunitinib (SUN) are drugs targeting vascular endothelial growth factor receptor 2 (VEGFR2). Sunitinib 39-48 kinase insert domain receptor Homo sapiens 75-120 30705246-7 2019 However, a greater-fold increase in sorafenib/sunitinib resistance was observed during hypoxia, particularly in pVHL-deficient cells. Sunitinib 46-55 von Hippel-Lindau tumor suppressor Homo sapiens 112-116 30689282-1 2019 Lenvatinib (LEN), sorafenib (SOR), and sunitinib (SUN) are drugs targeting vascular endothelial growth factor receptor 2 (VEGFR2). Sunitinib 39-48 kinase insert domain receptor Homo sapiens 122-128 30523507-4 2019 Second-line sunitinib potently inhibits KIT exon 13/14 mutants but is ineffective against exon 17 mutations. Sunitinib 12-21 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 40-43 30816432-0 2019 Potential tumor-suppressive role of microRNA-99a-3p in sunitinib-resistant renal cell carcinoma cells through the regulation of RRM2. Sunitinib 55-64 ribonucleotide reductase regulatory subunit M2 Homo sapiens 128-132 31083182-0 2019 Relationship between efficacy of sunitinib and KIT mutation of patients with advanced gastrointestinal stromal tumors after failure of imatinib: A systematic review. Sunitinib 33-42 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 47-50 31083182-3 2019 We initiated a systematic review to compare the efficacy of SU after failure of Imatinib (IM) in different KIT mutations. Sunitinib 60-62 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 107-110 31083182-9 2019 CONCLUSION: The results show that efficacy of SU is closely associated with KIT genotypes in GISTs. Sunitinib 46-48 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 76-79 30722031-2 2019 The first VEGF inhibitors approved for mRCC were sorafenib and sunitinib. Sunitinib 63-72 vascular endothelial growth factor A Homo sapiens 10-14 31013908-4 2019 Here, the antitumor effects of three conjugates possessing a selective binder of the extracellular portion of integrin alphaVbeta3 covalently linked to the tyrosine kinase inhibitor sunitinib were investigated in cisplatin-sensitive and -resistant ovarian carcinoma cells expressing both tyrosine kinase VEGFR2 and alphaVbeta3 at different levels. Sunitinib 182-191 integrin subunit alpha V Homo sapiens 110-130 30527746-0 2019 Fibroblast Growth Factor Receptor-2 Polymorphism rs2981582 is Correlated With Progression-free Survival and Overall Survival in Patients With Metastatic Clear-cell Renal Cell Carcinoma Treated With Sunitinib. Sunitinib 198-207 fibroblast growth factor receptor 2 Homo sapiens 0-35 30684595-2 2019 Sunitinib and regorafenib have been approved as the second and third line therapies to overcome some of these drug-resistance mutations; however, their limited clinical response, toxicity and resistance of the activation loop mutants still makes new therapies bearing different cKIT mutants activity spectrum profile highly demanded. Sunitinib 0-9 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 278-282 30984366-10 2019 Our data also support the hypothesis that the use of KIT/PDGFRA inhibitors, including non-approved agents, has improved OS for patients with imatinib- and sunitinib-resistant GIST. Sunitinib 155-164 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 53-56 30984366-10 2019 Our data also support the hypothesis that the use of KIT/PDGFRA inhibitors, including non-approved agents, has improved OS for patients with imatinib- and sunitinib-resistant GIST. Sunitinib 155-164 platelet derived growth factor receptor alpha Homo sapiens 57-63 30640790-6 2019 Sunitinib is an oral tyrosine kinase inhibitor that interacts with several angiogenesis receptors including platelet-derived growth factor receptors and VEGF receptors, and is approved for the first-line treatment in metastatic RCC. Sunitinib 0-9 vascular endothelial growth factor A Homo sapiens 153-157 30527746-2 2019 Recently, single nucleotide polymorphism (SNP) rs2981582 in Fibroblast Growth Factor Receptor 2 (FGFR2) was found to be associated with clinical outcome in patients with mccRCC treated with pazopanib and sunitinib. Sunitinib 204-213 fibroblast growth factor receptor 2 Homo sapiens 60-95 30527746-2 2019 Recently, single nucleotide polymorphism (SNP) rs2981582 in Fibroblast Growth Factor Receptor 2 (FGFR2) was found to be associated with clinical outcome in patients with mccRCC treated with pazopanib and sunitinib. Sunitinib 204-213 fibroblast growth factor receptor 2 Homo sapiens 97-102 30527746-12 2019 CONCLUSION: Polymorphism rs2981582 in FGFR2 is correlated to PFS and OS in patients with mccRCC treated with sunitinib. Sunitinib 109-118 fibroblast growth factor receptor 2 Homo sapiens 38-43 30527746-12 2019 CONCLUSION: Polymorphism rs2981582 in FGFR2 is correlated to PFS and OS in patients with mccRCC treated with sunitinib. Sunitinib 109-118 Moloney sarcoma oncogene Mus musculus 69-71 30792533-2 2019 Once resistance emerges, targeting KIT with tyrosine kinase inhibitors (TKIs) sunitinib and regorafenib provides clinical benefit, albeit of limited duration. Sunitinib 78-87 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 35-38 30689992-8 2019 Combination of sunitinib and TSA increased cell death with PARP cleavage, an early marker of mitochondrial apoptosis, whereas receptor tyrosine kinase signaling, which is the target of sunitinib, was not altered by TSA. Sunitinib 15-24 poly(ADP-ribose) polymerase 1 Homo sapiens 59-63 30620878-0 2019 Combination of Sunitinib and PD-L1 Blockade Enhances Anticancer Efficacy of TLR7/8 Agonist-Based Nanovaccine. Sunitinib 15-24 toll-like receptor 7 Mus musculus 76-82 30552230-5 2019 Here, we report that the ER-associated protein degradation (ERAD) inhibitor eeyarestatin sensitized MTC cells to the TKIs, sunitinib and vandetanib, thereby leading to synergistic upregulation of ATF4 expression, accumulation of reactive oxygen species, and subsequent cell death. Sunitinib 123-132 activating transcription factor 4 Homo sapiens 196-200 30821250-8 2019 Analysis of post-receptor pathways controlling proliferation and migration of HUVECs showed suppression of phosphorylated PI3K/Akt and ERK1/2 after exposure to sunitinib but not to 3PO in 10 microM concentration. Sunitinib 160-169 AKT serine/threonine kinase 1 Homo sapiens 127-130 30821250-8 2019 Analysis of post-receptor pathways controlling proliferation and migration of HUVECs showed suppression of phosphorylated PI3K/Akt and ERK1/2 after exposure to sunitinib but not to 3PO in 10 microM concentration. Sunitinib 160-169 mitogen-activated protein kinase 3 Homo sapiens 135-141 30693663-7 2019 Subsequent sunitinib (P = 0.066) and regorafenib (P = 0.003) were more commonly administered in period 2. Sunitinib 11-20 period circadian regulator 2 Homo sapiens 96-104 30779084-0 2019 Effect of miR-34a on resistance to sunitinib in breast cancer by regulating the Wnt/beta-catenin signaling pathway. Sunitinib 35-44 catenin beta 1 Homo sapiens 84-96 31031856-2 2019 Mammalian target of rapamycin (mTOR) inhibitor everolimus is currently used as a second-line therapy for sorafenib or sunitinib-refractory metastatic RCC patients. Sunitinib 118-127 mechanistic target of rapamycin kinase Homo sapiens 31-35 31031856-2 2019 Mammalian target of rapamycin (mTOR) inhibitor everolimus is currently used as a second-line therapy for sorafenib or sunitinib-refractory metastatic RCC patients. Sunitinib 118-127 mechanistic target of rapamycin kinase Homo sapiens 0-29 30401688-1 2019 PURPOSE: In the S-TRAC trial, adjuvant sunitinib prolonged disease-free survival (DFS) versus placebo in patients with loco-regional renal cell carcinoma at high risk of recurrence after nephrectomy. Sunitinib 39-48 T cell receptor alpha constant Homo sapiens 18-22 30401688-8 2019 Shorter DFS was observed for VEGFR1 rs9582036 C/A versus C/C with sunitinib, placebo, and combined therapies (P <= 0.05), and A/A versus C/C with sunitinib (P = 0.022). Sunitinib 66-75 fms related receptor tyrosine kinase 1 Homo sapiens 29-35 30401688-8 2019 Shorter DFS was observed for VEGFR1 rs9582036 C/A versus C/C with sunitinib, placebo, and combined therapies (P <= 0.05), and A/A versus C/C with sunitinib (P = 0.022). Sunitinib 149-158 fms related receptor tyrosine kinase 1 Homo sapiens 29-35 30401688-10 2019 CONCLUSIONS: Correlations between VEGFR1 and VEGFR2 SNPs and longer DFS with sunitinib suggest germline SNPs are predictive of improved outcomes with adjuvant sunitinib in patients with renal cell carcinoma. Sunitinib 77-86 fms related receptor tyrosine kinase 1 Homo sapiens 34-40 30401688-10 2019 CONCLUSIONS: Correlations between VEGFR1 and VEGFR2 SNPs and longer DFS with sunitinib suggest germline SNPs are predictive of improved outcomes with adjuvant sunitinib in patients with renal cell carcinoma. Sunitinib 77-86 kinase insert domain receptor Homo sapiens 45-51 30401688-10 2019 CONCLUSIONS: Correlations between VEGFR1 and VEGFR2 SNPs and longer DFS with sunitinib suggest germline SNPs are predictive of improved outcomes with adjuvant sunitinib in patients with renal cell carcinoma. Sunitinib 159-168 fms related receptor tyrosine kinase 1 Homo sapiens 34-40 30401688-10 2019 CONCLUSIONS: Correlations between VEGFR1 and VEGFR2 SNPs and longer DFS with sunitinib suggest germline SNPs are predictive of improved outcomes with adjuvant sunitinib in patients with renal cell carcinoma. Sunitinib 159-168 kinase insert domain receptor Homo sapiens 45-51 30648632-11 2019 CONCLUSIONS: Individualised sunitinib therapy is feasible, safe and an effective method to manage toxicity with one of the best efficacy seen for oral vascular endothelial growth factor inhibitors in metastatic renal cell carcinoma. Sunitinib 28-37 vascular endothelial growth factor A Homo sapiens 151-185 30779084-0 2019 Effect of miR-34a on resistance to sunitinib in breast cancer by regulating the Wnt/beta-catenin signaling pathway. Sunitinib 35-44 microRNA 34a Homo sapiens 10-17 30779084-0 2019 Effect of miR-34a on resistance to sunitinib in breast cancer by regulating the Wnt/beta-catenin signaling pathway. Sunitinib 35-44 Wnt family member 1 Homo sapiens 80-83 30779084-8 2019 RESULTS: The overexpression of miR-34a significantly reduced the invasive ability of MCF-7 cells after treatment with sunitinib. Sunitinib 118-127 microRNA 34a Homo sapiens 31-38 30779084-9 2019 After miR-34a expression was downregulated, the sensitivity of MCF-7 cells to sunitinib was significantly lowered. Sunitinib 78-87 microRNA 34a Homo sapiens 6-13 30779084-12 2019 CONCLUSIONS: MiR-34a affects the sensitivity to sunitinib in breast cancer by regulating the Wnt/beta-catenin signaling pathway. Sunitinib 48-57 microRNA 34a Homo sapiens 13-20 30779084-12 2019 CONCLUSIONS: MiR-34a affects the sensitivity to sunitinib in breast cancer by regulating the Wnt/beta-catenin signaling pathway. Sunitinib 48-57 Wnt family member 1 Homo sapiens 93-96 30779084-12 2019 CONCLUSIONS: MiR-34a affects the sensitivity to sunitinib in breast cancer by regulating the Wnt/beta-catenin signaling pathway. Sunitinib 48-57 catenin beta 1 Homo sapiens 97-109 30788170-6 2019 Sunitinib is required for KIT exon 9, 13, and 14 mutations, while ponatinib is used for exon 17 mutations and regorafenib for highly refractory tumors. Sunitinib 0-9 KIT proto-oncogene, receptor tyrosine kinase Canis lupus familiaris 26-29 28990511-1 2019 BACKGROUND: Sunitinib (SU11248) is an oral multi-target tyrosine kinase inhibitor (TKI) with low molecular weight, that inhibits platelet-derived growth factor receptors (PDGF-Rs) and vascular endothelial growth factor receptors (VEGFRs), c-KIT, fms-related tyrosine kinase 3 (FLT3) and RET. Sunitinib 12-21 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 239-244 30867824-12 2019 One of the most up-regulated genes in sunitinib-resistant cells was the CXCL5 cytokine. Sunitinib 38-47 C-X-C motif chemokine ligand 5 Homo sapiens 72-77 30867824-15 2019 The levels of CXCL5 present in the plasma of patients treated with sunitinib were predictive of the efficacy of sunitinib but not of the VEGF-directed antibody bevacizumab. Sunitinib 67-76 C-X-C motif chemokine ligand 5 Homo sapiens 14-19 30867824-15 2019 The levels of CXCL5 present in the plasma of patients treated with sunitinib were predictive of the efficacy of sunitinib but not of the VEGF-directed antibody bevacizumab. Sunitinib 112-121 C-X-C motif chemokine ligand 5 Homo sapiens 14-19 30498230-1 2019 BACKGROUND: There are phase 3 clinical trials underway evaluating anti-PD-L1 antibodies as adjuvant (postoperative) monotherapies for resectable renal cell carcinoma (RCC) and triple-negative breast cancer (TNBC); in combination with antiangiogenic VEGF/VEGFR2 inhibitors (e.g., bevacizumab and sunitinib) for metastatic RCC; and in combination with chemotherapeutics as neoadjuvant (preoperative) therapies for resectable TNBC. Sunitinib 295-304 CD274 antigen Mus musculus 71-76 30913495-0 2019 Elevated plasma interleukin 6 predicts poor response in patients treated with sunitinib for metastatic clear cell renal cell carcinoma. Sunitinib 78-87 interleukin 6 Homo sapiens 16-29 30566287-0 2019 Synthesis, Characterization and in vitro Studies of a Cathepsin B-Cleavable Prodrug of the VEGFR Inhibitor Sunitinib. Sunitinib 107-116 cathepsin B Homo sapiens 54-65 30566287-0 2019 Synthesis, Characterization and in vitro Studies of a Cathepsin B-Cleavable Prodrug of the VEGFR Inhibitor Sunitinib. Sunitinib 107-116 kinase insert domain receptor Homo sapiens 91-96 30566287-3 2019 Here, we present the synthesis and biological investigation of a cathepsin B-cleavable prodrug of the VEGFR inhibitor sunitinib. Sunitinib 118-127 cathepsin B Homo sapiens 65-76 30566287-3 2019 Here, we present the synthesis and biological investigation of a cathepsin B-cleavable prodrug of the VEGFR inhibitor sunitinib. Sunitinib 118-127 kinase insert domain receptor Homo sapiens 102-107 30854133-0 2019 Overexpression of FZD1 is Associated with a Good Prognosis and Resistance of Sunitinib in Clear Cell Renal Cell Carcinoma. Sunitinib 77-86 frizzled class receptor 1 Homo sapiens 18-22 30854133-5 2019 We then detected FZD1 mRNA expression in sunitinib-resistant cells and the corresponding parental cells by qRT-PCR. Sunitinib 41-50 frizzled class receptor 1 Homo sapiens 17-21 30854133-6 2019 FZD1 level was significantly upregulated in renal cancer tissues, renal cancer cell lines and their corresponding sunitinib-resistant cells. Sunitinib 114-123 frizzled class receptor 1 Homo sapiens 0-4 30854133-13 2019 In conclusion, our results suggested that expression status of FZD1 had a diagnostic value and prognostic value in ccRCC patients, it also may serve as a potential drug target to relieve sunitinib resistance in renal cancer patients. Sunitinib 187-196 frizzled class receptor 1 Homo sapiens 63-67 30719230-0 2019 ATF4 contributes to autophagy and survival in sunitinib treated brain tumor initiating cells (BTICs). Sunitinib 46-55 activating transcription factor 4 Homo sapiens 0-4 30719230-5 2019 We found that the ATF4/p-eIF2alpha pathway is activated in response to Sunitinib in primary tumor initiating progenitor cell cultures (BTICs). Sunitinib 71-80 activating transcription factor 4 Homo sapiens 18-22 30719230-5 2019 We found that the ATF4/p-eIF2alpha pathway is activated in response to Sunitinib in primary tumor initiating progenitor cell cultures (BTICs). Sunitinib 71-80 eukaryotic translation initiation factor 2A Homo sapiens 25-34 30719230-7 2019 In case of Sunitinib treated cells, autophagy is additionally increased by ATF4 mediated upregulation of autophagy genes. Sunitinib 11-20 activating transcription factor 4 Homo sapiens 75-79 30719230-8 2019 Inhibition of ATF4 by small interfering RNA (siRNA) reduced autophagy and cell proliferation after Sunitinib treatment in a subset of BTIC cultures. Sunitinib 99-108 activating transcription factor 4 Homo sapiens 14-18 30559346-5 2019 We additionally show, using preclinical mouse as well as patient data, that treatment with the inhibitor sunitinib significantly reduces the expression of INSR-A. Sunitinib 105-114 insulin receptor Homo sapiens 155-159 30073769-16 2019 CONCLUSIONS: Blocking of PlGF or injection of sunitinib results in a similar inhibition of neovascularization as by anti-VEGF treatment in the mouse model of ROP. Sunitinib 46-55 vascular endothelial growth factor A Mus musculus 121-125 30073769-17 2019 However, physiological angiogenesis that occurs after anti-VEGF treatment is blocked by anti-PlGF or sunitinib treatment, indicating that pathological neovascularization may follow different pathways than physiological angiogenesis. Sunitinib 101-110 vascular endothelial growth factor A Mus musculus 59-63 28990511-1 2019 BACKGROUND: Sunitinib (SU11248) is an oral multi-target tyrosine kinase inhibitor (TKI) with low molecular weight, that inhibits platelet-derived growth factor receptors (PDGF-Rs) and vascular endothelial growth factor receptors (VEGFRs), c-KIT, fms-related tyrosine kinase 3 (FLT3) and RET. Sunitinib 12-21 fms related receptor tyrosine kinase 3 Homo sapiens 246-275 28990511-1 2019 BACKGROUND: Sunitinib (SU11248) is an oral multi-target tyrosine kinase inhibitor (TKI) with low molecular weight, that inhibits platelet-derived growth factor receptors (PDGF-Rs) and vascular endothelial growth factor receptors (VEGFRs), c-KIT, fms-related tyrosine kinase 3 (FLT3) and RET. Sunitinib 12-21 fms related receptor tyrosine kinase 3 Homo sapiens 277-281 28990511-1 2019 BACKGROUND: Sunitinib (SU11248) is an oral multi-target tyrosine kinase inhibitor (TKI) with low molecular weight, that inhibits platelet-derived growth factor receptors (PDGF-Rs) and vascular endothelial growth factor receptors (VEGFRs), c-KIT, fms-related tyrosine kinase 3 (FLT3) and RET. Sunitinib 12-21 ret proto-oncogene Homo sapiens 287-290 28990511-1 2019 BACKGROUND: Sunitinib (SU11248) is an oral multi-target tyrosine kinase inhibitor (TKI) with low molecular weight, that inhibits platelet-derived growth factor receptors (PDGF-Rs) and vascular endothelial growth factor receptors (VEGFRs), c-KIT, fms-related tyrosine kinase 3 (FLT3) and RET. Sunitinib 23-30 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 239-244 28990511-1 2019 BACKGROUND: Sunitinib (SU11248) is an oral multi-target tyrosine kinase inhibitor (TKI) with low molecular weight, that inhibits platelet-derived growth factor receptors (PDGF-Rs) and vascular endothelial growth factor receptors (VEGFRs), c-KIT, fms-related tyrosine kinase 3 (FLT3) and RET. Sunitinib 23-30 fms related receptor tyrosine kinase 3 Homo sapiens 246-275 28990511-1 2019 BACKGROUND: Sunitinib (SU11248) is an oral multi-target tyrosine kinase inhibitor (TKI) with low molecular weight, that inhibits platelet-derived growth factor receptors (PDGF-Rs) and vascular endothelial growth factor receptors (VEGFRs), c-KIT, fms-related tyrosine kinase 3 (FLT3) and RET. Sunitinib 23-30 fms related receptor tyrosine kinase 3 Homo sapiens 277-281 28990511-1 2019 BACKGROUND: Sunitinib (SU11248) is an oral multi-target tyrosine kinase inhibitor (TKI) with low molecular weight, that inhibits platelet-derived growth factor receptors (PDGF-Rs) and vascular endothelial growth factor receptors (VEGFRs), c-KIT, fms-related tyrosine kinase 3 (FLT3) and RET. Sunitinib 23-30 ret proto-oncogene Homo sapiens 287-290 28990511-5 2019 RESULTS: In vitro studies showed that sunitinib targeted the cytosolic MEK/ERK and SAPK/JNK pathways in the RET/PTC1 cell inhibiting cell proliferation and causing stimulation of sodium/iodide symporter (NIS) gene expression in RET/PTC1 cells. Sunitinib 38-47 mitogen-activated protein kinase kinase 7 Homo sapiens 71-74 28990511-5 2019 RESULTS: In vitro studies showed that sunitinib targeted the cytosolic MEK/ERK and SAPK/JNK pathways in the RET/PTC1 cell inhibiting cell proliferation and causing stimulation of sodium/iodide symporter (NIS) gene expression in RET/PTC1 cells. Sunitinib 38-47 ret proto-oncogene Homo sapiens 108-111 28990511-5 2019 RESULTS: In vitro studies showed that sunitinib targeted the cytosolic MEK/ERK and SAPK/JNK pathways in the RET/PTC1 cell inhibiting cell proliferation and causing stimulation of sodium/iodide symporter (NIS) gene expression in RET/PTC1 cells. Sunitinib 38-47 patched 1 Homo sapiens 112-116 28990511-5 2019 RESULTS: In vitro studies showed that sunitinib targeted the cytosolic MEK/ERK and SAPK/JNK pathways in the RET/PTC1 cell inhibiting cell proliferation and causing stimulation of sodium/iodide symporter (NIS) gene expression in RET/PTC1 cells. Sunitinib 38-47 ret proto-oncogene Homo sapiens 228-231 28990511-5 2019 RESULTS: In vitro studies showed that sunitinib targeted the cytosolic MEK/ERK and SAPK/JNK pathways in the RET/PTC1 cell inhibiting cell proliferation and causing stimulation of sodium/iodide symporter (NIS) gene expression in RET/PTC1 cells. Sunitinib 38-47 patched 1 Homo sapiens 232-236 30281919-0 2019 NT5E inhibition suppresses the growth of sunitinib-resistant cells and EMT course and AKT/GSK-3beta signaling pathway in renal cell cancer. Sunitinib 41-50 5'-nucleotidase ecto Homo sapiens 0-4 30148679-8 2019 Functional studies confirm that E-cadherin is a key contributor to the survival of RCC cells under sunitinib treatment. Sunitinib 99-108 cadherin 1 Homo sapiens 32-42 30558998-7 2019 A positive correlation between gankyrin and sunitinib-resistance was also observed in RCC cell lines and in an orthotopic RCC model. Sunitinib 44-53 proteasome 26S subunit, non-ATPase 10 Homo sapiens 31-39 30281919-2 2019 Microarray analysis, quantitative real time PCR (qRT-PCR), Western blot, and immunohistochemistry were used to detect Ecto-5"-nucleotidase (NT5E) expressions in sunitinib-resistant tissues in RCC. Sunitinib 161-170 5'-nucleotidase ecto Homo sapiens 118-138 30281919-2 2019 Microarray analysis, quantitative real time PCR (qRT-PCR), Western blot, and immunohistochemistry were used to detect Ecto-5"-nucleotidase (NT5E) expressions in sunitinib-resistant tissues in RCC. Sunitinib 161-170 5'-nucleotidase ecto Homo sapiens 140-144 30281919-5 2019 In vivo experiment was performed using NCr-nu/nu mice to explore the effects of NT5E on cell growth and EMT signal in sunitinib environment. Sunitinib 118-127 5'-nucleotidase ecto Homo sapiens 80-84 30281919-6 2019 NT5E expression was upregulated in sunitinib-resistant RCC tissues and cells. Sunitinib 35-44 5'-nucleotidase ecto Homo sapiens 0-4 30281919-8 2019 EMT course and AKT/GSK-3beta signal pathway both in vitro and in vivo in sunitinib environment was suppressed to varying degrees via NT5E inhibition. Sunitinib 73-82 AKT serine/threonine kinase 1 Homo sapiens 15-18 30281919-8 2019 EMT course and AKT/GSK-3beta signal pathway both in vitro and in vivo in sunitinib environment was suppressed to varying degrees via NT5E inhibition. Sunitinib 73-82 glycogen synthase kinase 3 beta Homo sapiens 19-28 30281919-8 2019 EMT course and AKT/GSK-3beta signal pathway both in vitro and in vivo in sunitinib environment was suppressed to varying degrees via NT5E inhibition. Sunitinib 73-82 5'-nucleotidase ecto Homo sapiens 133-137 30281919-9 2019 NT5E inhibition could suppress the growth of sunitinib-resistant RCC cells and restrain EMT course and AKT/GSK-3beta signal pathway in sunitinib environment in RCC. Sunitinib 45-54 5'-nucleotidase ecto Homo sapiens 0-4 30281919-9 2019 NT5E inhibition could suppress the growth of sunitinib-resistant RCC cells and restrain EMT course and AKT/GSK-3beta signal pathway in sunitinib environment in RCC. Sunitinib 135-144 5'-nucleotidase ecto Homo sapiens 0-4 30281919-9 2019 NT5E inhibition could suppress the growth of sunitinib-resistant RCC cells and restrain EMT course and AKT/GSK-3beta signal pathway in sunitinib environment in RCC. Sunitinib 135-144 glycogen synthase kinase 3 beta Homo sapiens 107-116 31195398-7 2019 We evaluated the relationship between miR-130b and PTEN and also analyzed the effect of miR-130b on sunitinib resistance. Sunitinib 100-109 microRNA 130b Homo sapiens 88-96 29570259-11 2019 CONCLUSION: Our study suggests that dynamic changes in CRP can better predict survival in patients with mRCC who are treated with sunitinib. Sunitinib 130-139 C-reactive protein Homo sapiens 55-58 30653116-3 2019 We report the 1st case of metastatic jejunum GIST with a KIT exon 11 deletion that showed complete response (CR) to sunitinib for more than 3 years. Sunitinib 116-125 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 57-60 30653116-10 2019 LESSONS: In cases of metastatic jejunum GIST with a KIT exon 11 deletion, sunitinib as second-line therapy can be used to achieve CR for more than 3 years. Sunitinib 74-83 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 52-55 31195398-0 2019 miR-130b Promotes Sunitinib Resistance through Regulation of PTEN in Renal Cell Carcinoma. Sunitinib 18-27 microRNA 130b Homo sapiens 0-8 31195398-0 2019 miR-130b Promotes Sunitinib Resistance through Regulation of PTEN in Renal Cell Carcinoma. Sunitinib 18-27 phosphatase and tensin homolog Homo sapiens 61-65 31195398-4 2019 This study aimed to examine the expression and functional role of miR-130b and to analyze the association between miR-130b and sunitinib resistance in RCC. Sunitinib 127-136 microRNA 130b Homo sapiens 114-122 31195398-12 2019 Additionally, miR-130b was associated with sunitinib resistance through regulation of PTEN. Sunitinib 43-52 microRNA 130b Homo sapiens 14-22 31195398-12 2019 Additionally, miR-130b was associated with sunitinib resistance through regulation of PTEN. Sunitinib 43-52 phosphatase and tensin homolog Homo sapiens 86-90 31195398-13 2019 We established the sunitinib-resistant Caki-1 (Caki-1-SR) cells and observed that the expression of miR-130b was elevated in Caki-1-SR cells compared with parental Caki-1 cells. Sunitinib 19-28 microRNA 130b Homo sapiens 100-108 31195398-14 2019 Knockdown of miR-130b improved sunitinib resistance in Caki-1-SR cells. Sunitinib 31-40 microRNA 130b Homo sapiens 13-21 31195398-16 2019 miR-130b promoted cell growth and was associated with sunitinib resistance through regulating PTEN expression. Sunitinib 54-63 microRNA 130b Homo sapiens 0-8 31195398-16 2019 miR-130b promoted cell growth and was associated with sunitinib resistance through regulating PTEN expression. Sunitinib 54-63 phosphatase and tensin homolog Homo sapiens 94-98 30070687-5 2018 The impact of BCRP/ABCG2 on response to sunitinib treatment was investigated in two independent sunitinib-treated ccRCC-cohorts based on mRNA levels. Sunitinib 40-49 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 14-18 30393206-0 2019 Ageing alters the severity of Sunitinib-induced cardiotoxicity: Investigating the mitogen activated kinase kinase 7 pathway association. Sunitinib 30-39 mitogen activated protein kinase kinase 7 Rattus norvegicus 82-115 30393206-1 2019 Anti-cancer drug Sunitinib is linked to adverse cardiovascular events, which have shown to involve mitogen activated kinase kinase 7 (MKK7) pathway. Sunitinib 17-26 mitogen activated protein kinase kinase 7 Rattus norvegicus 99-132 30393206-1 2019 Anti-cancer drug Sunitinib is linked to adverse cardiovascular events, which have shown to involve mitogen activated kinase kinase 7 (MKK7) pathway. Sunitinib 17-26 mitogen activated protein kinase kinase 7 Rattus norvegicus 134-138 30393206-8 2019 Sunitinib treatment decreased the expression of miR-27a in all age groups, while miR-133a and miR-133b levels were increased in 3 months and decreased in 24 months groups. Sunitinib 0-9 microRNA 27a Rattus norvegicus 48-55 30393206-9 2019 MKK7 mRNA and p-MKK7 levels were decreased in the 3 months group after Sunitinib treatment. Sunitinib 71-80 mitogen activated protein kinase kinase 7 Rattus norvegicus 0-4 30393206-9 2019 MKK7 mRNA and p-MKK7 levels were decreased in the 3 months group after Sunitinib treatment. Sunitinib 71-80 mitogen activated protein kinase kinase 7 Rattus norvegicus 16-20 30393206-10 2019 MKK7 mRNA level was increased in 24 months group and p-MKK7 levels were increased in 12 months group following Sunitinib treatment. Sunitinib 111-120 mitogen activated protein kinase kinase 7 Rattus norvegicus 0-4 30393206-10 2019 MKK7 mRNA level was increased in 24 months group and p-MKK7 levels were increased in 12 months group following Sunitinib treatment. Sunitinib 111-120 mitogen activated protein kinase kinase 7 Rattus norvegicus 55-59 30393183-2 2018 In this study, our objective was to determine whether co-delivery of sunitinib, a member of tyrosine kinase inhibitor (TKI) family, and siRNA against Alox15 (siAlox15) could reverse the new-onset of T1D in non-obese diabetic (NOD)/ShiLtJ female mice which spontaneously develop diabetes. Sunitinib 69-78 arachidonate 15-lipoxygenase Mus musculus 150-156 30393183-4 2018 These micellar formulations exhibited sustained release of sunitinib for around 50% at 24 h and protected siAlox15 from serum degradation for at least 10 h. Further, both sunitinib and siAlox15 inhibited human peripheral blood mononuclear cell (PBMC) proliferation and activation by downregulating the phosphorylation of Akt, platelet-derived growth factor receptor (PDGFR) at protein levels and Alox15 in mRNA levels, respectively. Sunitinib 171-180 arachidonate 15-lipoxygenase Homo sapiens 108-114 30446580-9 2018 The patient received first-line sunitinib to target PDGFRA and PDGFRB and had stable disease for >6 mo, followed by nivolumab upon progression. Sunitinib 32-41 platelet derived growth factor receptor alpha Homo sapiens 52-58 30446580-9 2018 The patient received first-line sunitinib to target PDGFRA and PDGFRB and had stable disease for >6 mo, followed by nivolumab upon progression. Sunitinib 32-41 platelet derived growth factor receptor beta Homo sapiens 63-69 30070687-9 2018 BCRP/ABCG2 and MDR1/ABCB1 mRNA expression were linked to decreased progression-free survival after sunitinib treatment. Sunitinib 99-108 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 0-4 30070687-9 2018 BCRP/ABCG2 and MDR1/ABCB1 mRNA expression were linked to decreased progression-free survival after sunitinib treatment. Sunitinib 99-108 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 5-10 30070687-9 2018 BCRP/ABCG2 and MDR1/ABCB1 mRNA expression were linked to decreased progression-free survival after sunitinib treatment. Sunitinib 99-108 ATP binding cassette subfamily B member 1 Homo sapiens 15-19 30070687-9 2018 BCRP/ABCG2 and MDR1/ABCB1 mRNA expression were linked to decreased progression-free survival after sunitinib treatment. Sunitinib 99-108 ATP binding cassette subfamily B member 1 Homo sapiens 20-25 30070687-12 2018 In summary, BCRP/ABCG2 expression in ccRCC underlies considerable interindividual variability with impact on patient survival and response to sunitinib treatment. Sunitinib 142-151 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 12-16 30070687-12 2018 In summary, BCRP/ABCG2 expression in ccRCC underlies considerable interindividual variability with impact on patient survival and response to sunitinib treatment. Sunitinib 142-151 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 17-22 30369518-15 2018 And we found that 786-O RCC cells secrete high IL-6 levels after low dose stimulation with the TKIs sorafenib, sunitinib and pazopanib, inducing activation of AKT-mTOR pathway, NFkappaB, HIF-2alpha and VEGF expression. Sunitinib 111-120 AKT serine/threonine kinase 1 Homo sapiens 159-162 30651932-8 2018 Our findings suggest that axitinib works better than mTOR inhibitors after the first-line treatment with sunitinib. Sunitinib 105-114 mechanistic target of rapamycin kinase Homo sapiens 53-57 30651923-8 2018 Dynamic upward variations were also observed with respect to IL-8 levels in sunitinib-refractory individuals: 28.5 pg/mL at baseline vs. 38.3 pg/mL at 3 months, p-value=0.024. Sunitinib 76-85 C-X-C motif chemokine ligand 8 Homo sapiens 61-65 30651923-9 2018 In conclusion, two VEGFR-3 SNPs as well as various serum biomarkers were associated with diverse clinical outcomes in patients with well-differentiated pancreatic neuroendocrine tumors treated with sunitinib. Sunitinib 198-207 fms related receptor tyrosine kinase 4 Homo sapiens 19-26 30369518-15 2018 And we found that 786-O RCC cells secrete high IL-6 levels after low dose stimulation with the TKIs sorafenib, sunitinib and pazopanib, inducing activation of AKT-mTOR pathway, NFkappaB, HIF-2alpha and VEGF expression. Sunitinib 111-120 mechanistic target of rapamycin kinase Homo sapiens 163-167 30369518-15 2018 And we found that 786-O RCC cells secrete high IL-6 levels after low dose stimulation with the TKIs sorafenib, sunitinib and pazopanib, inducing activation of AKT-mTOR pathway, NFkappaB, HIF-2alpha and VEGF expression. Sunitinib 111-120 interleukin 6 Homo sapiens 47-51 30369518-15 2018 And we found that 786-O RCC cells secrete high IL-6 levels after low dose stimulation with the TKIs sorafenib, sunitinib and pazopanib, inducing activation of AKT-mTOR pathway, NFkappaB, HIF-2alpha and VEGF expression. Sunitinib 111-120 nuclear factor kappa B subunit 1 Homo sapiens 177-185 30369518-15 2018 And we found that 786-O RCC cells secrete high IL-6 levels after low dose stimulation with the TKIs sorafenib, sunitinib and pazopanib, inducing activation of AKT-mTOR pathway, NFkappaB, HIF-2alpha and VEGF expression. Sunitinib 111-120 endothelial PAS domain protein 1 Homo sapiens 187-197 30369518-15 2018 And we found that 786-O RCC cells secrete high IL-6 levels after low dose stimulation with the TKIs sorafenib, sunitinib and pazopanib, inducing activation of AKT-mTOR pathway, NFkappaB, HIF-2alpha and VEGF expression. Sunitinib 111-120 vascular endothelial growth factor A Homo sapiens 202-206 30267660-0 2018 RCAN1.4 acts as a suppressor of cancer progression and sunitinib resistance in clear cell renal cell carcinoma. Sunitinib 55-64 regulator of calcineurin 1 Homo sapiens 0-5 28782406-0 2018 Severe sunitinib-induced myelosuppression in a patient with a CYP 3A4 polymorphism. Sunitinib 7-16 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 62-69 30514344-4 2018 In this review, we summarized the preclinical and clinical studies on new FLT3 inhibitors, including sorafenib, lestaurtinib, sunitinib, tandutinib, quizartinib, midostaurin, gilteritinib, crenolanib, cabozantinib, Sel24-B489, G-749, AMG 925, TTT-3002, and FF-10101. Sunitinib 126-135 fms related receptor tyrosine kinase 3 Homo sapiens 74-78 30267660-8 2018 In addition, RCAN1.4 expression was downregulated in sunitinib-resistant renal cancer cell lines, and inhibition of RCAN1.4 promoted sunitinib resistance. Sunitinib 53-62 regulator of calcineurin 1 Homo sapiens 13-18 30267660-8 2018 In addition, RCAN1.4 expression was downregulated in sunitinib-resistant renal cancer cell lines, and inhibition of RCAN1.4 promoted sunitinib resistance. Sunitinib 133-142 regulator of calcineurin 1 Homo sapiens 116-121 30267660-9 2018 We also found that RCAN1.4 could regulate epithelial-mesenchymal transition (EMT) and the expression of HIF2alpha in sunitinib-resistant cell lines. Sunitinib 117-126 regulator of calcineurin 1 Homo sapiens 19-24 30267660-9 2018 We also found that RCAN1.4 could regulate epithelial-mesenchymal transition (EMT) and the expression of HIF2alpha in sunitinib-resistant cell lines. Sunitinib 117-126 endothelial PAS domain protein 1 Homo sapiens 104-113 30267660-10 2018 Taken together, these findings indicate that downregulation of RCAN1.4 may be crucial for the metastasis of ccRCC and may induce sunitinib resistance. Sunitinib 129-138 regulator of calcineurin 1 Homo sapiens 63-68 30428867-11 2018 CONCLUSIONS: Sunitinib is a multi-targeted inhibitor of vascular endothelial growth factor (VEGF) receptors. Sunitinib 13-22 vascular endothelial growth factor A Homo sapiens 56-90 30428867-11 2018 CONCLUSIONS: Sunitinib is a multi-targeted inhibitor of vascular endothelial growth factor (VEGF) receptors. Sunitinib 13-22 vascular endothelial growth factor A Homo sapiens 92-96 30348622-0 2018 Screening of FDA-approved drugs identifies sutent as a modulator of UCP1 expression in brown adipose tissue. Sunitinib 43-49 uncoupling protein 1 (mitochondrial, proton carrier) Mus musculus 68-72 30713805-7 2019 Sunitinib improved pericyte coverage on endothelial cells and the expression levels of regulator of G-protein signaling 5, suggesting blood vessel normalization. Sunitinib 0-9 regulator of G protein signaling 5 Homo sapiens 87-121 30419914-0 2018 Sunitinib-suppressed miR-452-5p facilitates renal cancer cell invasion and metastasis through modulating SMAD4/SMAD7 signals. Sunitinib 0-9 microRNA 452 Homo sapiens 21-28 30419914-0 2018 Sunitinib-suppressed miR-452-5p facilitates renal cancer cell invasion and metastasis through modulating SMAD4/SMAD7 signals. Sunitinib 0-9 SMAD family member 4 Homo sapiens 105-110 30419914-0 2018 Sunitinib-suppressed miR-452-5p facilitates renal cancer cell invasion and metastasis through modulating SMAD4/SMAD7 signals. Sunitinib 0-9 SMAD family member 7 Homo sapiens 111-116 30419914-3 2018 EXPERIMENTAL DESIGN: We focused on 2 published microarray data to select out our anchored miRNA and then explored the roles of miR-452-5p both in vitro and in vivo, which was downregulated after Sunitinib treatment while upregulated in metastasis renal cell carcinoma (RCC) tissues. Sunitinib 195-204 microRNA 452 Homo sapiens 127-134 30419914-4 2018 RESULTS: Here, we discovered that treating with Sunitinib, the targeted receptor tyrosine kinase inhibitor (TKI), inhibited renal cancer cell migration and invasion via attenuating the expression of miR-452-5p. Sunitinib 48-57 microRNA 4525 Homo sapiens 199-209 30055290-0 2018 Targeting autophagy by small molecule inhibitors of vacuolar protein sorting 34 (Vps34) improves the sensitivity of breast cancer cells to Sunitinib. Sunitinib 139-148 phosphatidylinositol 3-kinase catalytic subunit type 3 Homo sapiens 81-86 30372397-7 2018 Because 3p loss and VHL inactivation are nearly universal truncal events in ccRCC, the resulting HIF1/2 signaling overdrive and accompanied tumor hypervascularization probably underlie the therapeutic benefits observed with vascular endothelial growth factor receptor inhibitors, including sorafenib, sunitinib, pazopanib, axitinib, bevacizumab, cabozantinib, and lenvatinib. Sunitinib 301-310 SET domain containing 2, histone lysine methyltransferase Homo sapiens 97-101 30031043-5 2018 Sunitinib did not affect body weight, but increased plasma ALT activity 6-fold. Sunitinib 0-9 glutamic pyruvic transaminase, soluble Mus musculus 59-62 30031043-10 2018 Protein expression of PGC-1alpha, citrate synthase activity and mtDNA copy number were all decreased in livers of sunitinib-treated mice, indicating impaired mitochondrial proliferation. Sunitinib 114-123 peroxisome proliferative activated receptor, gamma, coactivator 1 alpha Mus musculus 22-32 30031043-10 2018 Protein expression of PGC-1alpha, citrate synthase activity and mtDNA copy number were all decreased in livers of sunitinib-treated mice, indicating impaired mitochondrial proliferation. Sunitinib 114-123 citrate synthase Mus musculus 34-50 30031043-11 2018 Caspase 3 activation and TUNEL-positive hepatocytes were increased in livers of sunitinib-treated mice, indicating hepatocyte apoptosis. Sunitinib 80-89 caspase 3 Mus musculus 0-9 30055290-0 2018 Targeting autophagy by small molecule inhibitors of vacuolar protein sorting 34 (Vps34) improves the sensitivity of breast cancer cells to Sunitinib. Sunitinib 139-148 phosphatidylinositol 3-kinase catalytic subunit type 3 Homo sapiens 52-79 30055290-7 2018 Vps34 inhibitor significantly potentiated cytotoxicity of Sunitinib and Erlotinib in MCF-7 and MDA-MB-231 in vitro in monolayer cultures and when grown as multicellular spheroids. Sunitinib 58-67 phosphatidylinositol 3-kinase catalytic subunit type 3 Homo sapiens 0-5 30071205-7 2018 Treatment of mice with sunitinib, a VEGF receptor tyrosine kinase inhibitor, once (day 2) or twice (days 1 and 2) post-infection reduced mortality by 50-80% compared with untreated mice. Sunitinib 23-32 vascular endothelial growth factor A Mus musculus 36-40 30071205-8 2018 Notably, sunitinib treatment decreased serum TNF levels, white blood cell counts, and hematocrit levels relative to untreated mice, but had only marginal effects on tissue viral burden. Sunitinib 9-18 tumor necrosis factor Mus musculus 45-48 30412222-2 2018 We report safety, therapy management, and patient-reported outcomes for patients receiving sunitinib and placebo in the S-TRAC trial. Sunitinib 91-100 T cell receptor alpha constant Homo sapiens 122-126 29896907-9 2018 Sunitinib +ABCC2 blocker group showed a significant response to therapy compared to the other treatment groups both in vitro (P <= 0.0001) and in vivo (P = 0.0132). Sunitinib 0-9 ATP binding cassette subfamily C member 2 Homo sapiens 11-16 29896907-10 2018 ABCC2 blockage resulted in higher sunitinib uptake, both in vitro (P = 0.0016) and in vivo (P = 0.0031). Sunitinib 34-43 ATP binding cassette subfamily C member 2 Homo sapiens 0-5 30524898-0 2018 Sphingosine kinase 1 overexpression contributes to sunitinib resistance in clear cell renal cell carcinoma. Sunitinib 51-60 sphingosine kinase 1 Homo sapiens 0-20 30524898-8 2018 Our results unraveled that increased SphK1 kinase activation defines an important mechanism for sunitinib resistance, therefore contributes to tumour development and represents therapeutic targets for ccRCC. Sunitinib 96-105 sphingosine kinase 1 Homo sapiens 37-42 30200486-0 2018 Effects of Sunitinib and Other Kinase Inhibitors on Cells Harboring a PDGFRB Mutation Associated with Infantile Myofibromatosis. Sunitinib 11-20 platelet derived growth factor receptor beta Homo sapiens 70-76 30371202-0 2018 Renal Soluble Guanylate Cyclase Is Downregulated in Sunitinib-Induced Hypertension. Sunitinib 52-61 guanylate cyclase 1 soluble subunit alpha 1 Rattus norvegicus 6-31 29983397-9 2018 Sunitinib inhibited Complex I with an IC50 value of 38 microM and caused ATP depletion, caspase 3/7 activation, an increase in reactive oxygen species (ROS), and decreased nucleoside and glucose uptake. Sunitinib 0-9 caspase 3 Mus musculus 88-97 30194077-7 2018 In renal clear cell carcinoma, the target FLT1 of pazopanib, sunitinib, sorafenib, and axitinib was recurrently deleted, whereas FLT4 bound by the same drugs, was recurrently amplified. Sunitinib 61-70 fms related receptor tyrosine kinase 1 Homo sapiens 42-46 29764854-2 2018 Treatment with the small-molecule inhibitors imatinib, sunitinib, and regorafenib resulted in resistance (c-KIT mutant tumors) or limited activity (c-KIT wild-type tumors). Sunitinib 55-64 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 106-111 29764854-2 2018 Treatment with the small-molecule inhibitors imatinib, sunitinib, and regorafenib resulted in resistance (c-KIT mutant tumors) or limited activity (c-KIT wild-type tumors). Sunitinib 55-64 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 148-153 30371202-9 2018 Renal cortical sGC mRNA and sGC alpha1-subunit protein abundance were less in sunitinib-treated rats than in controls, and cinaciguat effectively lowered arterial pressure by 15-20 mm Hg in sunitinib-treated rats. Sunitinib 78-87 guanylate cyclase 1 soluble subunit alpha 1 Rattus norvegicus 15-18 30371202-10 2018 Conclusions Renal cortical sGC is downregulated in the presence of intact endothelium-dependent renal vascular resistance regulation in developing sunitinib-induced hypertension. Sunitinib 147-156 guanylate cyclase 1 soluble subunit alpha 1 Rattus norvegicus 27-30 30371202-11 2018 This suggests that sGC downregulation occurs outside the renal vasculature, increases renal sodium retention, and contributes to nitrate resistance of sunitinib-induced hypertension. Sunitinib 151-160 guanylate cyclase 1 soluble subunit alpha 1 Rattus norvegicus 19-22 30200486-7 2018 We also observed that PDGFR-beta phosphorylation in tumor cells is reduced by the receptor tyrosine kinase inhibitor sunitinib. Sunitinib 117-126 platelet derived growth factor receptor beta Homo sapiens 22-32 30200486-8 2018 In contrast, MAPK/ERK kinases (MEK) 1/2 and ERK1/2 kinases remained constitutively phosphorylated after treatment with sunitinib and other relevant protein kinase inhibitors. Sunitinib 119-128 mitogen-activated protein kinase kinase 1 Homo sapiens 18-39 30200486-8 2018 In contrast, MAPK/ERK kinases (MEK) 1/2 and ERK1/2 kinases remained constitutively phosphorylated after treatment with sunitinib and other relevant protein kinase inhibitors. Sunitinib 119-128 mitogen-activated protein kinase 3 Homo sapiens 44-50 30200486-9 2018 Our study showed that sunitinib is a very promising agent that affects the proliferation of tumor cells with a p.R561C mutation in PDGFR-beta. Sunitinib 22-31 platelet derived growth factor receptor beta Homo sapiens 131-141 30008308-4 2018 Therefore, a novel linkable sunitinib derivative, designated SB1, was rationally designed and synthesized. Sunitinib 28-37 Sh3kbp1 binding protein 1 Mus musculus 61-64 30224936-1 2018 Objectives: Sunitinib (a second-line chemotherapeutic agent that inhibits multiple kinases, including KIT and PDGFR) is widely used in imatinib-resistant patients with gastrointestinal stromal tumors (GISTs). Sunitinib 12-21 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 102-105 30224936-1 2018 Objectives: Sunitinib (a second-line chemotherapeutic agent that inhibits multiple kinases, including KIT and PDGFR) is widely used in imatinib-resistant patients with gastrointestinal stromal tumors (GISTs). Sunitinib 12-21 platelet derived growth factor receptor beta Homo sapiens 110-115 30224936-8 2018 Patients with KIT mutations showed an improved CBR to sunitinib compared to those with PDGFRA mutations. Sunitinib 54-63 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 14-17 30008308-7 2018 Cytotoxicity evaluation in three glioma cells showed that SB1 preserved the antiproliferative effect of sunitinib. Sunitinib 104-113 Sh3kbp1 binding protein 1 Mus musculus 58-61 29803017-0 2018 Hypoxia-Targeting Drug Evofosfamide (TH-302) Enhances Sunitinib Activity in Neuroblastoma Xenograft Models. Sunitinib 54-63 Rho guanine nucleotide exchange factor (GEF) 16 Mus musculus 76-89 30109169-9 2018 IL-8 was increased in sunitinib-resistant Caki-2 and SN12K1 cells and decreased in 786-0 without any significant changes in Caki-1. Sunitinib 22-31 C-X-C motif chemokine ligand 8 Homo sapiens 0-4 30109169-12 2018 The increased IL-6 may contribute to sunitinib resistance either via VEGF-mediated angiogenesis or through shifting of the Bcl2/Bax balance in favour of anti-apoptosis. Sunitinib 37-46 interleukin 6 Homo sapiens 14-18 30109169-12 2018 The increased IL-6 may contribute to sunitinib resistance either via VEGF-mediated angiogenesis or through shifting of the Bcl2/Bax balance in favour of anti-apoptosis. Sunitinib 37-46 vascular endothelial growth factor A Homo sapiens 69-73 30109169-12 2018 The increased IL-6 may contribute to sunitinib resistance either via VEGF-mediated angiogenesis or through shifting of the Bcl2/Bax balance in favour of anti-apoptosis. Sunitinib 37-46 BCL2 apoptosis regulator Homo sapiens 123-127 30109169-12 2018 The increased IL-6 may contribute to sunitinib resistance either via VEGF-mediated angiogenesis or through shifting of the Bcl2/Bax balance in favour of anti-apoptosis. Sunitinib 37-46 BCL2 associated X, apoptosis regulator Homo sapiens 128-131 29935772-2 2018 Sunitinib, a multikinase inhibitor, was the first Fms-like tyrosine kinase 3 (FLT3) inhibitor clinically used against AML. Sunitinib 0-9 fms related receptor tyrosine kinase 3 Homo sapiens 50-76 29935772-2 2018 Sunitinib, a multikinase inhibitor, was the first Fms-like tyrosine kinase 3 (FLT3) inhibitor clinically used against AML. Sunitinib 0-9 fms related receptor tyrosine kinase 3 Homo sapiens 78-82 29986726-15 2018 Sunitinib, gemcitabine and chloroquine treated mice showed a significant reduction of GRP78 expression, reduced cell proliferation and increased apoptosis in pancreas, compatible with a tumor response. Sunitinib 0-9 heat shock protein 5 Mus musculus 86-91 29963235-2 2018 We aimed to establish molecular imaging during treatment with sunitinib using the fibronectin extradomain A specific small immunoprotein(SIP)-F8 in glioma. Sunitinib 62-71 fibronectin 1 Mus musculus 82-93 29729400-5 2018 Motherwort total alkaloids significantly increased angiogenesis in transgenic Tg (flk1: EGFP) zebrafish embryos treated with sunitinib, as did stachydrine, the most abundant alkaloid produced by motherwort. Sunitinib 125-134 kinase insert domain receptor (a type III receptor tyrosine kinase) Danio rerio 82-86 29704617-10 2018 Based upon the X-ray crystallographic structures, imatinib, sunitinib, and ponatinib are Type II Kit inhibitors. Sunitinib 60-69 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 97-100 29915248-8 2018 Gefitinib, imatinib, pazopanib, sorafenib, and sunitinib also inhibited MATE1-mediated creatinine uptake. Sunitinib 47-56 solute carrier family 47 member 1 Rattus norvegicus 72-77 29396852-5 2018 Then the in vitro data also demonstrated the efficacy of Sunitinib, a currently used drug to treat ccRCC, could be increased after targeting this newly identified miR-32-5p/TR4/HGF/Met signaling. Sunitinib 57-66 microRNA 32 Homo sapiens 163-169 29396852-5 2018 Then the in vitro data also demonstrated the efficacy of Sunitinib, a currently used drug to treat ccRCC, could be increased after targeting this newly identified miR-32-5p/TR4/HGF/Met signaling. Sunitinib 57-66 nuclear receptor subfamily 2 group C member 2 Homo sapiens 173-176 29396852-5 2018 Then the in vitro data also demonstrated the efficacy of Sunitinib, a currently used drug to treat ccRCC, could be increased after targeting this newly identified miR-32-5p/TR4/HGF/Met signaling. Sunitinib 57-66 hepatocyte growth factor Homo sapiens 177-180 29678462-2 2018 Clinically used kinase inhibitors imatinib and sunitinib are potent inhibitors of wild-type PDGFR family members, but show reduced binding to mutant forms. Sunitinib 47-56 platelet derived growth factor receptor beta Homo sapiens 92-97 29572225-6 2018 Additionally, AR overexpression resulted in acquired sunitinib resistance and the AR antagonist enzalutamide-induced AR degradation and attenuated AR downstream activity in sunitinib-resistant cells, also indicated by decreased secretion of human kallikrein 2. Sunitinib 173-182 androgen receptor Homo sapiens 14-16 29877179-6 2018 In contrast, the S-TRAC trial showed improved DFS after one year of adjuvant sunitinib using central review process, but not using investigator review process. Sunitinib 77-86 T cell receptor alpha constant Homo sapiens 19-23 29572225-0 2018 Therapeutic Targeting of Sunitinib-Induced AR Phosphorylation in Renal Cell Carcinoma. Sunitinib 25-34 androgen receptor Homo sapiens 43-45 29572225-3 2018 Through integrative analysis of a reverse phase protein array, we discovered increased expression of AR in an RCC patient-derived xenograft model of acquired resistance to the receptor tyrosine kinase inhibitor (RTKi) sunitinib. Sunitinib 218-227 androgen receptor Homo sapiens 101-103 29572225-6 2018 Additionally, AR overexpression resulted in acquired sunitinib resistance and the AR antagonist enzalutamide-induced AR degradation and attenuated AR downstream activity in sunitinib-resistant cells, also indicated by decreased secretion of human kallikrein 2. Sunitinib 173-182 androgen receptor Homo sapiens 82-84 29572225-4 2018 AR expression was increased in RCC cell lines with either acquired or intrinsic sunitinib resistance in vitro An AR signaling gene array profiler indicated elevated levels of AR target genes in sunitinib-resistant cells. Sunitinib 80-89 androgen receptor Homo sapiens 0-2 29572225-4 2018 AR expression was increased in RCC cell lines with either acquired or intrinsic sunitinib resistance in vitro An AR signaling gene array profiler indicated elevated levels of AR target genes in sunitinib-resistant cells. Sunitinib 194-203 androgen receptor Homo sapiens 0-2 29572225-6 2018 Additionally, AR overexpression resulted in acquired sunitinib resistance and the AR antagonist enzalutamide-induced AR degradation and attenuated AR downstream activity in sunitinib-resistant cells, also indicated by decreased secretion of human kallikrein 2. Sunitinib 173-182 androgen receptor Homo sapiens 82-84 29572225-4 2018 AR expression was increased in RCC cell lines with either acquired or intrinsic sunitinib resistance in vitro An AR signaling gene array profiler indicated elevated levels of AR target genes in sunitinib-resistant cells. Sunitinib 194-203 androgen receptor Homo sapiens 113-115 29572225-4 2018 AR expression was increased in RCC cell lines with either acquired or intrinsic sunitinib resistance in vitro An AR signaling gene array profiler indicated elevated levels of AR target genes in sunitinib-resistant cells. Sunitinib 194-203 androgen receptor Homo sapiens 113-115 29572225-6 2018 Additionally, AR overexpression resulted in acquired sunitinib resistance and the AR antagonist enzalutamide-induced AR degradation and attenuated AR downstream activity in sunitinib-resistant cells, also indicated by decreased secretion of human kallikrein 2. Sunitinib 173-182 androgen receptor Homo sapiens 82-84 29572225-5 2018 Sunitinib-induced AR transcriptional activity was associated with increased phosphorylation of serine 81 (pS81) on AR. Sunitinib 0-9 androgen receptor Homo sapiens 18-20 29572225-6 2018 Additionally, AR overexpression resulted in acquired sunitinib resistance and the AR antagonist enzalutamide-induced AR degradation and attenuated AR downstream activity in sunitinib-resistant cells, also indicated by decreased secretion of human kallikrein 2. Sunitinib 173-182 kallikrein related peptidase 2 Homo sapiens 247-259 29572225-5 2018 Sunitinib-induced AR transcriptional activity was associated with increased phosphorylation of serine 81 (pS81) on AR. Sunitinib 0-9 androgen receptor Homo sapiens 115-117 29572225-6 2018 Additionally, AR overexpression resulted in acquired sunitinib resistance and the AR antagonist enzalutamide-induced AR degradation and attenuated AR downstream activity in sunitinib-resistant cells, also indicated by decreased secretion of human kallikrein 2. Sunitinib 53-62 androgen receptor Homo sapiens 14-16 29490989-0 2018 A Genetic Polymorphism in CTLA-4 Is Associated with Overall Survival in Sunitinib-Treated Patients with Clear Cell Metastatic Renal Cell Carcinoma. Sunitinib 72-81 cytotoxic T-lymphocyte associated protein 4 Homo sapiens 26-32 28874077-0 2018 Effects of sunitinib and bevacizumab on VEGF and miRNA levels on corneal neovascularization. Sunitinib 11-20 vascular endothelial growth factor A Rattus norvegicus 40-44 28874077-9 2018 Also, in comparison with the control group; VEGF-A expression was downregulated by nearly 0.75 times in sunitinib group and nearly 0.52 times in bevacizumab group. Sunitinib 104-113 vascular endothelial growth factor A Rattus norvegicus 44-50 28874077-10 2018 VEGFR-2 expression was downregulated by 0.89 times in sunitinib group and 0.68 times in bevacizumab group, compared to the control group. Sunitinib 54-63 kinase insert domain receptor Rattus norvegicus 0-7 28874077-11 2018 miR-15 b, miR-16 and miR-126 levels were statistically lower in sunitinib and bevacizumab groups, but miR-188 and miR-410 levels were two-fold higher compared to the control group. Sunitinib 64-73 microRNA 15b Rattus norvegicus 0-8 28874077-12 2018 The miR-210 level was found higher only in sunitinib group compared to the control group. Sunitinib 43-52 microRNA 210 Rattus norvegicus 4-11 28874077-14 2018 CONCLUSION: Topical application of bevacizumab (5 mg/ml) and sunitinib (0.5 mg/ml) decreases the levels of VEGFR-2 and VEGF-A in CNV. Sunitinib 61-70 kinase insert domain receptor Rattus norvegicus 107-114 28874077-14 2018 CONCLUSION: Topical application of bevacizumab (5 mg/ml) and sunitinib (0.5 mg/ml) decreases the levels of VEGFR-2 and VEGF-A in CNV. Sunitinib 61-70 vascular endothelial growth factor A Rattus norvegicus 119-125 29633072-0 2018 Association of Expression Levels or Activation Status of STAT3 with Treatment Outcomes of Sunitinib in Patients with Renal Cell Carcinoma. Sunitinib 90-99 signal transducer and activator of transcription 3 Homo sapiens 57-62 29633072-2 2018 OBJECTIVE: This retrospective study aimed to elucidate the association of STAT3 expression in tumor cells with the therapeutic outcomes of sunitinib in patients with renal cell carcinoma (RCC). Sunitinib 139-148 signal transducer and activator of transcription 3 Homo sapiens 74-79 29633072-10 2018 CONCLUSIONS: Activated STAT3 in tumor tissues shows a significant association with poor prognosis in patients with RCC who received sunitinib as a first-line therapy, and positive p-STAT3 expression could be a potential biomarker for refractoriness to sunitinib therapy. Sunitinib 132-141 signal transducer and activator of transcription 3 Homo sapiens 23-28 29633072-10 2018 CONCLUSIONS: Activated STAT3 in tumor tissues shows a significant association with poor prognosis in patients with RCC who received sunitinib as a first-line therapy, and positive p-STAT3 expression could be a potential biomarker for refractoriness to sunitinib therapy. Sunitinib 252-261 signal transducer and activator of transcription 3 Homo sapiens 23-28 29490989-8 2018 No significant associations with PFS were found.Conclusions: The G-allele of rs231775 in the CTLA-4 gene is associated with an improved OS in sunitinib-treated cc-mRCC patients and could potentially be used as a prognostic biomarker. Sunitinib 142-151 cytotoxic T-lymphocyte associated protein 4 Homo sapiens 93-99 30202791-3 2018 The second wave of tyrosine kinase inhibitors (TKIs), which target the intracellular site of VEGF receptor kinases, began with the approval of sorafenib in 2005 and sunitinib in 2006. Sunitinib 165-174 vascular endothelial growth factor A Homo sapiens 93-97 29382533-2 2018 Sunitinib, a multi-targeted tyrosine kinase inhibitor, is known to selectively inhibit several growth factor receptors, including vascular endothelial growth factor receptor, platelet-derived growth factor receptor and stem cell factor. Sunitinib 0-9 KIT ligand Homo sapiens 219-235 29532289-7 2018 RESULTS: In vitro, sunitinib and sorafenib significantly inhibited platelet aggregation (20 muM sunitinib: 71.3%, p < 0.001; 25 muM sorafenib: 55.8%, p = 0.042). Sunitinib 19-28 latexin Homo sapiens 92-95 29532289-7 2018 RESULTS: In vitro, sunitinib and sorafenib significantly inhibited platelet aggregation (20 muM sunitinib: 71.3%, p < 0.001; 25 muM sorafenib: 55.8%, p = 0.042). Sunitinib 19-28 latexin Homo sapiens 131-134 29532289-8 2018 Sorafenib and sunitinib significantly inhibited P-selectin expression on platelets. Sunitinib 14-23 selectin P Homo sapiens 48-58 29760553-4 2018 The immunohistochemistry in this patient revealed abundant expression of platelet-derived growth factor receptor-beta on tumor cells, which is one of the drug targets of sunitinib. Sunitinib 170-179 platelet derived growth factor receptor beta Homo sapiens 73-117 29796168-4 2018 Here, we aimed to elucidate the anti-cancer effects of JQ1 and the mechanisms underlying BRD4 inhibition in sunitinib-sensitive and -resistant ccRCCs. Sunitinib 108-117 bromodomain containing 4 Homo sapiens 89-93 29796168-8 2018 Chromatin immunoprecipitation assays revealed that these oncogenes may be promising BRD4 targets, particularly in sunitinib-resistant ccRCC cells. Sunitinib 114-123 bromodomain containing 4 Homo sapiens 84-88 29796168-9 2018 These results identified SCG5, SPOCD1, RGS19, and ARHGAP22 as potential prognostic markers and showed that BRD4 inhibition may have applications as a potential therapeutic approach in sunitinib-sensitive and -resistant ccRCC. Sunitinib 184-193 bromodomain containing 4 Homo sapiens 107-111 29854307-8 2018 Moreover, TRC105 enhanced the inhibitory effect of Sunitinib on VEGF signaling and reduced VEGFR2-Akt-Creb activation, suggesting a molecular cooperation between the two drugs. Sunitinib 51-60 vascular endothelial growth factor A Homo sapiens 64-68 29563634-0 2018 The tyrosine-kinase inhibitor sunitinib targets vascular endothelial (VE)-cadherin: a marker of response to antitumoural treatment in metastatic renal cell carcinoma. Sunitinib 30-39 cadherin 5 Homo sapiens 48-82 29563634-7 2018 RESULTS: Human VE-cadherin is a direct target for sunitinib which inhibits its VEGF-induced phosphorylation and cleavage on endothelial monolayer and endothelial cell migration in the 3D model. Sunitinib 50-59 cadherin 5 Homo sapiens 15-26 29563634-7 2018 RESULTS: Human VE-cadherin is a direct target for sunitinib which inhibits its VEGF-induced phosphorylation and cleavage on endothelial monolayer and endothelial cell migration in the 3D model. Sunitinib 50-59 vascular endothelial growth factor A Homo sapiens 79-83 29854303-0 2018 The role of netrin-1 in metastatic renal cell carcinoma treated with sunitinib. Sunitinib 69-78 netrin 1 Homo sapiens 12-20 29854303-7 2018 NTN1 was assessed in our in vivo sunitinib-conditioned mouse model using immunostaining. Sunitinib 33-42 netrin 1 Mus musculus 0-4 29854303-9 2018 Results: Human renal cell carcinoma sunitinib-conditioned cell lines showed upregulation of netrin-1 in microarray and q-PCR. Sunitinib 36-45 netrin 1 Homo sapiens 92-100 29854303-11 2018 Silencing of netrin-1 in sunitinib-conditioned Caki-1 cells did not demonstrate a significant reduction in cell migration. Sunitinib 25-34 netrin 1 Homo sapiens 13-21 29854303-12 2018 Conclusion: Netrin-1 is highly upregulated in renal cell carcinoma treated with sunitinib, but has no influence on cell viability or cell migration in metastatic RCC. Sunitinib 80-89 netrin 1 Homo sapiens 12-20 29854307-7 2018 At a molecular level, we showed that the combination of TRC105 and Sunitinib induced the phosphorylation of Smad 2/3 to promote endothelial cell death. Sunitinib 67-76 SMAD family member 2 Homo sapiens 108-116 29382533-5 2018 We found that Sunitinib could inhibit the proliferation of Hacat/E cell in a time and concentration dependent manner by influencing the expression level of cell cycle protein D1, cycle protein E1, in addition, Sunitinib could induce the apoptosis of Hacat/E cell and up-regulate the expression of poly ADP-ribose polymerase (PARP). Sunitinib 14-23 poly(ADP-ribose) polymerase 1 Homo sapiens 297-323 29382533-5 2018 We found that Sunitinib could inhibit the proliferation of Hacat/E cell in a time and concentration dependent manner by influencing the expression level of cell cycle protein D1, cycle protein E1, in addition, Sunitinib could induce the apoptosis of Hacat/E cell and up-regulate the expression of poly ADP-ribose polymerase (PARP). Sunitinib 14-23 poly(ADP-ribose) polymerase 1 Homo sapiens 325-329 29382533-5 2018 We found that Sunitinib could inhibit the proliferation of Hacat/E cell in a time and concentration dependent manner by influencing the expression level of cell cycle protein D1, cycle protein E1, in addition, Sunitinib could induce the apoptosis of Hacat/E cell and up-regulate the expression of poly ADP-ribose polymerase (PARP). Sunitinib 210-219 poly(ADP-ribose) polymerase 1 Homo sapiens 297-323 29382533-5 2018 We found that Sunitinib could inhibit the proliferation of Hacat/E cell in a time and concentration dependent manner by influencing the expression level of cell cycle protein D1, cycle protein E1, in addition, Sunitinib could induce the apoptosis of Hacat/E cell and up-regulate the expression of poly ADP-ribose polymerase (PARP). Sunitinib 210-219 poly(ADP-ribose) polymerase 1 Homo sapiens 325-329 29374054-1 2018 Purpose: Adjuvant sunitinib therapy compared with placebo prolonged disease-free survival (DFS) in patients with locoregional high-risk renal cell carcinoma (RCC) in the S-TRAC trial (ClinicalTrials.gov number NCT00375674). Sunitinib 18-27 T cell receptor alpha constant Homo sapiens 172-176 29095068-1 2018 OBJECTIVES: Clear-cell renal cell carcinomas (ccRCC) are characterized by hyper-vascularization and can respond to vascular endothelial growth factor receptor (VEGFR) inhibitors such as sunitinib. Sunitinib 186-195 kinase insert domain receptor Homo sapiens 115-158 29095068-1 2018 OBJECTIVES: Clear-cell renal cell carcinomas (ccRCC) are characterized by hyper-vascularization and can respond to vascular endothelial growth factor receptor (VEGFR) inhibitors such as sunitinib. Sunitinib 186-195 kinase insert domain receptor Homo sapiens 160-165 29095068-9 2018 HIF2A, VEGFA, VEGFR1, VEGFR2 and VEGFR3 were highly expressed in the transcriptomic ccrcc2-subtype of tumors, known to be highly sensitive to sunitinib. Sunitinib 142-151 endothelial PAS domain protein 1 Homo sapiens 0-5 29095068-9 2018 HIF2A, VEGFA, VEGFR1, VEGFR2 and VEGFR3 were highly expressed in the transcriptomic ccrcc2-subtype of tumors, known to be highly sensitive to sunitinib. Sunitinib 142-151 vascular endothelial growth factor A Homo sapiens 7-12 29095068-9 2018 HIF2A, VEGFA, VEGFR1, VEGFR2 and VEGFR3 were highly expressed in the transcriptomic ccrcc2-subtype of tumors, known to be highly sensitive to sunitinib. Sunitinib 142-151 fms related receptor tyrosine kinase 1 Homo sapiens 14-20 29095068-9 2018 HIF2A, VEGFA, VEGFR1, VEGFR2 and VEGFR3 were highly expressed in the transcriptomic ccrcc2-subtype of tumors, known to be highly sensitive to sunitinib. Sunitinib 142-151 kinase insert domain receptor Homo sapiens 22-28 29095068-9 2018 HIF2A, VEGFA, VEGFR1, VEGFR2 and VEGFR3 were highly expressed in the transcriptomic ccrcc2-subtype of tumors, known to be highly sensitive to sunitinib. Sunitinib 142-151 fms related receptor tyrosine kinase 4 Homo sapiens 33-39 29095068-12 2018 CONCLUSIONS: Intratumoral expression of genes involved in the HIF-VEGF-VEGFR-pro-angiogenic pathway, especially VEGFR2, is associated with favorable outcome on sunitinib in m-ccRCCs. Sunitinib 160-169 vascular endothelial growth factor A Homo sapiens 66-70 29095068-12 2018 CONCLUSIONS: Intratumoral expression of genes involved in the HIF-VEGF-VEGFR-pro-angiogenic pathway, especially VEGFR2, is associated with favorable outcome on sunitinib in m-ccRCCs. Sunitinib 160-169 kinase insert domain receptor Homo sapiens 71-76 29095068-12 2018 CONCLUSIONS: Intratumoral expression of genes involved in the HIF-VEGF-VEGFR-pro-angiogenic pathway, especially VEGFR2, is associated with favorable outcome on sunitinib in m-ccRCCs. Sunitinib 160-169 kinase insert domain receptor Homo sapiens 112-118 29374054-5 2018 Among patients with IHC, longer DFS was observed in patients with tumor CD8+ T-cell density >= versus < median [median (95% CI), not reached (6.83-not reached) versus 3.47 years (1.73-not reached); hazard ratio (HR) 0.40 (95% CI, 0.20-0.81); P = 0.009] treated with sunitinib (n = 101), but not with placebo (n = 90). Sunitinib 272-281 CD8a molecule Homo sapiens 72-75 29374054-8 2018 Among all patients with PD-L1+ tumors, DFS was numerically longer with sunitinib versus placebo (HR 0.58; P = 0.175).Conclusions: Greater CD8+ T-cell density in tumor tissue was associated with longer DFS with sunitinib but not placebo, suggesting predictive treatment effect utility. Sunitinib 71-80 CD8a molecule Homo sapiens 138-141 29374054-8 2018 Among all patients with PD-L1+ tumors, DFS was numerically longer with sunitinib versus placebo (HR 0.58; P = 0.175).Conclusions: Greater CD8+ T-cell density in tumor tissue was associated with longer DFS with sunitinib but not placebo, suggesting predictive treatment effect utility. Sunitinib 210-219 CD8a molecule Homo sapiens 138-141 29532881-12 2018 By contrast, the sunitinib-adapted cells exhibited increased neuropilin-1 (NRP1) expression levels compared with the control cells. Sunitinib 17-26 neuropilin 1 Homo sapiens 61-73 29532881-12 2018 By contrast, the sunitinib-adapted cells exhibited increased neuropilin-1 (NRP1) expression levels compared with the control cells. Sunitinib 17-26 neuropilin 1 Homo sapiens 75-79 29532881-13 2018 In the sunitinib-adapted cells, NRP1 interacted with phosphorylated cMet, and the cMet activation was dependent on NRP1. Sunitinib 7-16 neuropilin 1 Homo sapiens 32-36 29532881-13 2018 In the sunitinib-adapted cells, NRP1 interacted with phosphorylated cMet, and the cMet activation was dependent on NRP1. Sunitinib 7-16 MET proto-oncogene, receptor tyrosine kinase Homo sapiens 68-72 29532881-13 2018 In the sunitinib-adapted cells, NRP1 interacted with phosphorylated cMet, and the cMet activation was dependent on NRP1. Sunitinib 7-16 MET proto-oncogene, receptor tyrosine kinase Homo sapiens 82-86 29532881-13 2018 In the sunitinib-adapted cells, NRP1 interacted with phosphorylated cMet, and the cMet activation was dependent on NRP1. Sunitinib 7-16 neuropilin 1 Homo sapiens 115-119 29532881-14 2018 Thus, NRP1 or cMet blockade suppressed the evasive activation of the sunitinib-adapted cells. Sunitinib 69-78 neuropilin 1 Homo sapiens 6-10 29532881-14 2018 Thus, NRP1 or cMet blockade suppressed the evasive activation of the sunitinib-adapted cells. Sunitinib 69-78 MET proto-oncogene, receptor tyrosine kinase Homo sapiens 14-18 29532881-15 2018 These results suggest that the sunitinib-adapted cells switched from a VEGF-R-dependent pathway to an alternative NRP1/cMet-dependent one. Sunitinib 31-40 vascular endothelial growth factor A Homo sapiens 71-75 29532881-15 2018 These results suggest that the sunitinib-adapted cells switched from a VEGF-R-dependent pathway to an alternative NRP1/cMet-dependent one. Sunitinib 31-40 neuropilin 1 Homo sapiens 114-118 29532881-15 2018 These results suggest that the sunitinib-adapted cells switched from a VEGF-R-dependent pathway to an alternative NRP1/cMet-dependent one. Sunitinib 31-40 MET proto-oncogene, receptor tyrosine kinase Homo sapiens 119-123 29554654-9 2018 This article reviews the literature behind the preclinical and clinical data of the combination of RT with VEGFR-TKIs currently approved for RCC (sunitinib, sorafenib, pazopanib, and axitinib), with a focus on dose schedules as well as efficacy and toxicity. Sunitinib 146-155 kinase insert domain receptor Homo sapiens 107-112 29283974-9 2018 After 14 days of treatment, renal medullary beta-ENaC protein abundance was higher in rats that received sunitinib than in controls, whereas alpha-ENaC, gamma-ENaC, and NCC abundances were similar in both groups. Sunitinib 105-114 sodium channel epithelial 1 subunit beta Rattus norvegicus 44-53 29665322-9 2018 Low tumour cell expression of IL6Ralpha was significantly associated with improved response to sunitinib (Fisher"s exact test, p = 0.03), but not with PFS or OS. Sunitinib 95-104 interleukin 6 receptor Homo sapiens 30-39 29665322-13 2018 Loss of tumour cell expression of IL6Ralpha in mccRCC patients treated with sunitinib predicts improved treatment response, and might represent a candidate predictive marker. Sunitinib 76-85 interleukin 6 receptor Homo sapiens 34-43 29721091-0 2018 Soluble CD146 is a predictive marker of pejorative evolution and of sunitinib efficacy in clear cell renal cell carcinoma. Sunitinib 68-77 melanoma cell adhesion molecule Homo sapiens 8-13 29721091-7 2018 In vitro, resistant cells to sunitinib expressed high levels of CD146 mRNA and protein in comparison to sensitive cells. Sunitinib 29-38 melanoma cell adhesion molecule Homo sapiens 64-69 29721091-8 2018 Moreover, recombinant CD146 protected cells from the sunitinib-dependent decrease of cell viability. Sunitinib 53-62 melanoma cell adhesion molecule Homo sapiens 22-27 29721091-9 2018 Conclusion: CD146/sCD146 produced by tumor cells is a relevant biological marker of ccRCC aggressiveness and relapse on sunitinib treatment. Sunitinib 120-129 melanoma cell adhesion molecule Homo sapiens 12-17 29615901-2 2018 It was hypothesized that sunitinib-induced fatigue may be related to off target inhibition of the AMPK enzyme, which results in impairment of energy-producing processes at a systemic level. Sunitinib 25-34 protein kinase AMP-activated non-catalytic subunit beta 1 Homo sapiens 98-102 29356495-3 2018 To address tractability of multiple RTKs for chemical degradation by the E3 ligase CUL4-RBX1-DDB1-CRBN (CRL4CRBN), we synthesized a series of phthalimide degraders based on the promiscuous kinase inhibitors sunitinib and PHA665752. Sunitinib 207-216 damage specific DNA binding protein 1 Homo sapiens 93-97 29356495-3 2018 To address tractability of multiple RTKs for chemical degradation by the E3 ligase CUL4-RBX1-DDB1-CRBN (CRL4CRBN), we synthesized a series of phthalimide degraders based on the promiscuous kinase inhibitors sunitinib and PHA665752. Sunitinib 207-216 cereblon Homo sapiens 98-102 29376753-9 2018 Left ventricular systolic dysfunction was increased after tyrosine kinase inhibitor therapy (relative risk = 2.53; 95% confidence interval:1.79 - 3.57; p < 0.001), with the highest risks reported for sunitinib and hepatocellular cancer. Sunitinib 203-212 TXK tyrosine kinase Homo sapiens 58-73 29515108-0 2018 The convergent roles of NF-kappaB and ER stress in sunitinib-mediated expression of pro-tumorigenic cytokines and refractory phenotype in renal cell carcinoma. Sunitinib 51-60 nuclear factor kappa B subunit 1 Homo sapiens 24-33 29515108-4 2018 Our studies reveal that sunitinib triggers two resistance-promoting signaling pathways in RCC cells, which emanate from the endoplasmic reticulum (ER) stress response: a PERK-driven ER stress response that induces expression of the pro-tumorigenic cytokines IL-6, IL-8, and TNF-alpha, and a TRAF2-mediated NF-kappaB survival program that protects tumor cells against cell death. Sunitinib 24-33 eukaryotic translation initiation factor 2 alpha kinase 3 Homo sapiens 170-174 29515108-4 2018 Our studies reveal that sunitinib triggers two resistance-promoting signaling pathways in RCC cells, which emanate from the endoplasmic reticulum (ER) stress response: a PERK-driven ER stress response that induces expression of the pro-tumorigenic cytokines IL-6, IL-8, and TNF-alpha, and a TRAF2-mediated NF-kappaB survival program that protects tumor cells against cell death. Sunitinib 24-33 interleukin 6 Homo sapiens 258-262 29515108-4 2018 Our studies reveal that sunitinib triggers two resistance-promoting signaling pathways in RCC cells, which emanate from the endoplasmic reticulum (ER) stress response: a PERK-driven ER stress response that induces expression of the pro-tumorigenic cytokines IL-6, IL-8, and TNF-alpha, and a TRAF2-mediated NF-kappaB survival program that protects tumor cells against cell death. Sunitinib 24-33 C-X-C motif chemokine ligand 8 Homo sapiens 264-268 29515108-4 2018 Our studies reveal that sunitinib triggers two resistance-promoting signaling pathways in RCC cells, which emanate from the endoplasmic reticulum (ER) stress response: a PERK-driven ER stress response that induces expression of the pro-tumorigenic cytokines IL-6, IL-8, and TNF-alpha, and a TRAF2-mediated NF-kappaB survival program that protects tumor cells against cell death. Sunitinib 24-33 tumor necrosis factor Homo sapiens 274-283 29515108-4 2018 Our studies reveal that sunitinib triggers two resistance-promoting signaling pathways in RCC cells, which emanate from the endoplasmic reticulum (ER) stress response: a PERK-driven ER stress response that induces expression of the pro-tumorigenic cytokines IL-6, IL-8, and TNF-alpha, and a TRAF2-mediated NF-kappaB survival program that protects tumor cells against cell death. Sunitinib 24-33 TNF receptor associated factor 2 Homo sapiens 291-296 29515108-4 2018 Our studies reveal that sunitinib triggers two resistance-promoting signaling pathways in RCC cells, which emanate from the endoplasmic reticulum (ER) stress response: a PERK-driven ER stress response that induces expression of the pro-tumorigenic cytokines IL-6, IL-8, and TNF-alpha, and a TRAF2-mediated NF-kappaB survival program that protects tumor cells against cell death. Sunitinib 24-33 nuclear factor kappa B subunit 1 Homo sapiens 306-315 29515108-5 2018 PERK blockade completely prevents sunitinib-induced expression of IL-6, IL-8 and TNF-alpha, whereas NF-kappaB inhibition reinstates sensitivity of RCC cells to sunitinib both in vitro and in vivo. Sunitinib 34-43 eukaryotic translation initiation factor 2 alpha kinase 3 Homo sapiens 0-4 29515108-5 2018 PERK blockade completely prevents sunitinib-induced expression of IL-6, IL-8 and TNF-alpha, whereas NF-kappaB inhibition reinstates sensitivity of RCC cells to sunitinib both in vitro and in vivo. Sunitinib 34-43 interleukin 6 Homo sapiens 66-70 29515108-5 2018 PERK blockade completely prevents sunitinib-induced expression of IL-6, IL-8 and TNF-alpha, whereas NF-kappaB inhibition reinstates sensitivity of RCC cells to sunitinib both in vitro and in vivo. Sunitinib 34-43 C-X-C motif chemokine ligand 8 Homo sapiens 72-76 29515108-5 2018 PERK blockade completely prevents sunitinib-induced expression of IL-6, IL-8 and TNF-alpha, whereas NF-kappaB inhibition reinstates sensitivity of RCC cells to sunitinib both in vitro and in vivo. Sunitinib 34-43 tumor necrosis factor Homo sapiens 81-90 29248607-0 2018 Involvement of mitogen activated kinase kinase 7 intracellular signalling pathway in Sunitinib-induced cardiotoxicity. Sunitinib 85-94 mitogen-activated protein kinase kinase 7 Homo sapiens 15-48 29435010-5 2018 The inhibition of autophagy by CQ enhanced sunitinib-induced apoptosis, which was characterized by the activation of caspase-3, caspase-9, Bcl-2 and p53. Sunitinib 43-52 caspase 3 Homo sapiens 117-126 29435010-5 2018 The inhibition of autophagy by CQ enhanced sunitinib-induced apoptosis, which was characterized by the activation of caspase-3, caspase-9, Bcl-2 and p53. Sunitinib 43-52 caspase 9 Homo sapiens 128-137 29435010-5 2018 The inhibition of autophagy by CQ enhanced sunitinib-induced apoptosis, which was characterized by the activation of caspase-3, caspase-9, Bcl-2 and p53. Sunitinib 43-52 BCL2 apoptosis regulator Homo sapiens 139-144 29435010-5 2018 The inhibition of autophagy by CQ enhanced sunitinib-induced apoptosis, which was characterized by the activation of caspase-3, caspase-9, Bcl-2 and p53. Sunitinib 43-52 tumor protein p53 Homo sapiens 149-152 29435010-6 2018 Additionally, the exposure of OS-RC-2 cells to CQ and sunitinib resulted in the inhibition of AKT, tuberous sclerosis complex 2, mechanistic target of rapamycin and p70 ribosomal S6 kinase, which are associated with cell proliferation. Sunitinib 54-63 AKT serine/threonine kinase 1 Homo sapiens 94-97 29345296-9 2018 Among the test agents, sunitinib and cisplatin decreased the secretion of vascular endothelial growth factor (VEGF)-A from the A549 lung cancer cells. Sunitinib 23-32 vascular endothelial growth factor A Homo sapiens 74-108 29345296-9 2018 Among the test agents, sunitinib and cisplatin decreased the secretion of vascular endothelial growth factor (VEGF)-A from the A549 lung cancer cells. Sunitinib 23-32 vascular endothelial growth factor A Homo sapiens 110-114 29248607-7 2018 Administration of Sunitinib (1 muM) during Langendorff perfusion resulted in increased infarct size, increased miR-133a expression, and decreased phosphorylation of the ASK1/MKK7/JNK pathway compared to control. Sunitinib 18-27 mitogen-activated protein kinase 8 Homo sapiens 179-182 29248607-8 2018 Co-administration of NQDI-1 (2.5 muM) attenuated the increased Sunitinib-induced infarct size, reversed miR-133a expression and restored phosphorylated levels of ASK1/MKK7/JNK. Sunitinib 63-72 mitogen-activated protein kinase kinase 7 Homo sapiens 167-171 29248607-8 2018 Co-administration of NQDI-1 (2.5 muM) attenuated the increased Sunitinib-induced infarct size, reversed miR-133a expression and restored phosphorylated levels of ASK1/MKK7/JNK. Sunitinib 63-72 mitogen-activated protein kinase 8 Homo sapiens 172-175 29248607-4 2018 This study investigates the role of ASK1, MKK7 and JNK during Sunitinib-induced cardiotoxicity. Sunitinib 62-71 mitogen-activated protein kinase kinase kinase 5 Homo sapiens 36-40 29248607-9 2018 These findings suggest that the ASK1/MKK7/JNK intracellular signalling pathway is important in Sunitinib-induced cardiotoxicity. Sunitinib 95-104 mitogen-activated protein kinase kinase kinase 5 Homo sapiens 32-36 29248607-9 2018 These findings suggest that the ASK1/MKK7/JNK intracellular signalling pathway is important in Sunitinib-induced cardiotoxicity. Sunitinib 95-104 mitogen-activated protein kinase kinase 7 Homo sapiens 37-41 29248607-9 2018 These findings suggest that the ASK1/MKK7/JNK intracellular signalling pathway is important in Sunitinib-induced cardiotoxicity. Sunitinib 95-104 mitogen-activated protein kinase 8 Homo sapiens 42-45 29248607-13 2018 Investigations into the ASK1/MKK7/JNK transduction pathway could lead to development of cardioprotective adjunct therapy, which could prevent Sunitinib-induced cardiac injury. Sunitinib 142-151 mitogen-activated protein kinase kinase kinase 5 Homo sapiens 24-28 29248607-13 2018 Investigations into the ASK1/MKK7/JNK transduction pathway could lead to development of cardioprotective adjunct therapy, which could prevent Sunitinib-induced cardiac injury. Sunitinib 142-151 mitogen-activated protein kinase kinase 7 Homo sapiens 29-33 29248607-13 2018 Investigations into the ASK1/MKK7/JNK transduction pathway could lead to development of cardioprotective adjunct therapy, which could prevent Sunitinib-induced cardiac injury. Sunitinib 142-151 mitogen-activated protein kinase 8 Homo sapiens 34-37 29248607-4 2018 This study investigates the role of ASK1, MKK7 and JNK during Sunitinib-induced cardiotoxicity. Sunitinib 62-71 mitogen-activated protein kinase 8 Homo sapiens 51-54 29248607-7 2018 Administration of Sunitinib (1 muM) during Langendorff perfusion resulted in increased infarct size, increased miR-133a expression, and decreased phosphorylation of the ASK1/MKK7/JNK pathway compared to control. Sunitinib 18-27 mitogen-activated protein kinase kinase kinase 5 Homo sapiens 169-173 29248607-7 2018 Administration of Sunitinib (1 muM) during Langendorff perfusion resulted in increased infarct size, increased miR-133a expression, and decreased phosphorylation of the ASK1/MKK7/JNK pathway compared to control. Sunitinib 18-27 mitogen-activated protein kinase kinase 7 Homo sapiens 174-178 29551130-6 2018 The first TK to be targeted, Bcr-Abl, led to the generation of several drugs including imatinib, dasatinib, and sunitinib that provided a rich understanding of this phenomenon. Sunitinib 112-121 ABL proto-oncogene 1, non-receptor tyrosine kinase Homo sapiens 29-36 28676023-1 2018 BACKGROUND: Sunitinib, a tyrosine kinase inhibitor of vascular endothelial growth factor (VEGF), is approved for first and second line treatment of advanced renal cell carcinoma (RCC). Sunitinib 12-21 vascular endothelial growth factor A Homo sapiens 54-88 28676023-1 2018 BACKGROUND: Sunitinib, a tyrosine kinase inhibitor of vascular endothelial growth factor (VEGF), is approved for first and second line treatment of advanced renal cell carcinoma (RCC). Sunitinib 12-21 vascular endothelial growth factor A Homo sapiens 90-94 29662541-2 2018 Chronic treatment with vascular endothelial growth factor receptor (VEGFR)-targeting sunitinib upregulates MET and AXL in RCC, indicating that cabozantinib may be particularly effective in patients with advanced RCC whose disease progressed on prior VEGFR-targeted treatment. Sunitinib 85-94 kinase insert domain receptor Homo sapiens 23-66 29662541-2 2018 Chronic treatment with vascular endothelial growth factor receptor (VEGFR)-targeting sunitinib upregulates MET and AXL in RCC, indicating that cabozantinib may be particularly effective in patients with advanced RCC whose disease progressed on prior VEGFR-targeted treatment. Sunitinib 85-94 kinase insert domain receptor Homo sapiens 68-73 29662541-2 2018 Chronic treatment with vascular endothelial growth factor receptor (VEGFR)-targeting sunitinib upregulates MET and AXL in RCC, indicating that cabozantinib may be particularly effective in patients with advanced RCC whose disease progressed on prior VEGFR-targeted treatment. Sunitinib 85-94 AXL receptor tyrosine kinase Homo sapiens 115-118 29662541-2 2018 Chronic treatment with vascular endothelial growth factor receptor (VEGFR)-targeting sunitinib upregulates MET and AXL in RCC, indicating that cabozantinib may be particularly effective in patients with advanced RCC whose disease progressed on prior VEGFR-targeted treatment. Sunitinib 85-94 kinase insert domain receptor Homo sapiens 250-255 29298329-12 2018 In addition, 48 h sutent treatment significantly down-regulated the protein expression of PDGFRbeta, MYCN, SOX2 and Survivin in the AI tumorspheres and inhibited tumorsphere self-renewal. Sunitinib 18-24 platelet derived growth factor receptor beta Homo sapiens 90-99 29298329-12 2018 In addition, 48 h sutent treatment significantly down-regulated the protein expression of PDGFRbeta, MYCN, SOX2 and Survivin in the AI tumorspheres and inhibited tumorsphere self-renewal. Sunitinib 18-24 MYCN proto-oncogene, bHLH transcription factor Homo sapiens 101-105 29298329-12 2018 In addition, 48 h sutent treatment significantly down-regulated the protein expression of PDGFRbeta, MYCN, SOX2 and Survivin in the AI tumorspheres and inhibited tumorsphere self-renewal. Sunitinib 18-24 SRY-box transcription factor 2 Homo sapiens 107-111 29581831-6 2018 Results: Necessary dose reductions of sunitinib were significantly correlated with SNP rs1933437 in FLT3. Sunitinib 38-47 fms related receptor tyrosine kinase 3 Homo sapiens 100-104 29581831-9 2018 Our study provides a population PK model of sunitinib with the ABCB1 genotype as a predictive covariate for apparent oral clearance. Sunitinib 44-53 ATP binding cassette subfamily B member 1 Homo sapiens 63-68 30205729-0 2018 HMGB1 represses the anti-cancer activity of sunitinib by governing TP53 autophagic degradation via its nucleus-to-cytoplasm transport. Sunitinib 44-53 high mobility group box 1 Homo sapiens 0-5 30205729-0 2018 HMGB1 represses the anti-cancer activity of sunitinib by governing TP53 autophagic degradation via its nucleus-to-cytoplasm transport. Sunitinib 44-53 tumor protein p53 Homo sapiens 67-71 30205729-3 2018 Here we report that resistance to sunitinib therapy was driven by autophagic degradation of TP53/p53. Sunitinib 34-43 tumor protein p53 Homo sapiens 92-96 30205729-3 2018 Here we report that resistance to sunitinib therapy was driven by autophagic degradation of TP53/p53. Sunitinib 34-43 tumor protein p53 Homo sapiens 97-100 30205729-5 2018 Mechanistically, the transport of TP53 from the nucleus to the cytoplasm was essential for the sunitinib-induced autophagic degradation of TP53 and did not require TP53 nuclear export signals (NESs). Sunitinib 95-104 tumor protein p53 Homo sapiens 34-38 30205729-5 2018 Mechanistically, the transport of TP53 from the nucleus to the cytoplasm was essential for the sunitinib-induced autophagic degradation of TP53 and did not require TP53 nuclear export signals (NESs). Sunitinib 95-104 tumor protein p53 Homo sapiens 139-143 30205729-5 2018 Mechanistically, the transport of TP53 from the nucleus to the cytoplasm was essential for the sunitinib-induced autophagic degradation of TP53 and did not require TP53 nuclear export signals (NESs). Sunitinib 95-104 tumor protein p53 Homo sapiens 139-143 30205729-7 2018 The inhibition of HMGB1 sensitized cancer cells to sunitinib. Sunitinib 51-60 high mobility group box 1 Homo sapiens 18-23 30205729-8 2018 Importantly, sunitinib induced the degradation of all TP53 proteins, except for TP53 proteins with mutations in the interaction domain of TP53 with HMGB1 (amino acids 313 to 352). Sunitinib 13-22 tumor protein p53 Homo sapiens 54-58 30205729-9 2018 In conclusion, our data identify an alternative HMGB1-mediated TP53 protein turnover mechanism that participates in the resistance of sunitinib and suggest HMGB1 as a potential therapeutic target for improving clinical outcomes of sunitinib. Sunitinib 134-143 high mobility group box 1 Homo sapiens 48-53 30205729-9 2018 In conclusion, our data identify an alternative HMGB1-mediated TP53 protein turnover mechanism that participates in the resistance of sunitinib and suggest HMGB1 as a potential therapeutic target for improving clinical outcomes of sunitinib. Sunitinib 134-143 tumor protein p53 Homo sapiens 63-67 30205729-9 2018 In conclusion, our data identify an alternative HMGB1-mediated TP53 protein turnover mechanism that participates in the resistance of sunitinib and suggest HMGB1 as a potential therapeutic target for improving clinical outcomes of sunitinib. Sunitinib 231-240 high mobility group box 1 Homo sapiens 48-53 30205729-9 2018 In conclusion, our data identify an alternative HMGB1-mediated TP53 protein turnover mechanism that participates in the resistance of sunitinib and suggest HMGB1 as a potential therapeutic target for improving clinical outcomes of sunitinib. Sunitinib 231-240 tumor protein p53 Homo sapiens 63-67 30205729-9 2018 In conclusion, our data identify an alternative HMGB1-mediated TP53 protein turnover mechanism that participates in the resistance of sunitinib and suggest HMGB1 as a potential therapeutic target for improving clinical outcomes of sunitinib. Sunitinib 231-240 high mobility group box 1 Homo sapiens 156-161 29161241-10 2018 A 10% reduction in marker lesions was associated with improved PFS in the whole sunitinib population (HR 0.55 (95 CI 0.3-0.9); P=0.04); mostly in patients on sunitinib CDD (HR 0.33 (95% CI 0.2-0.7); P=0.005). Sunitinib 80-89 natriuretic peptide A Homo sapiens 168-171 29652183-0 2018 Effects of sunitinib on immunoreactivity of vimentin, E-cadherin and S100 in kidneys of streptozotocin induced diabetic mice. Sunitinib 11-20 vimentin Mus musculus 44-52 29652183-14 2018 We found that the number of vimentin and E-cadherin positive glomeruli and tubules were increased after sunitinib treatment compared to saline treated diabetic mice. Sunitinib 104-113 vimentin Mus musculus 28-36 29652183-14 2018 We found that the number of vimentin and E-cadherin positive glomeruli and tubules were increased after sunitinib treatment compared to saline treated diabetic mice. Sunitinib 104-113 cadherin 1 Mus musculus 41-51 29652183-15 2018 The number of vimentin labeled tubules was decreased in the sunitinib treated group compared to diabetic + saline groups. Sunitinib 60-69 vimentin Mus musculus 14-22 29652183-17 2018 The effect of sunitinib on experimental diabetic mice appears to be related to levels of vimentin, E-cadherin and S100 in the glomeruli and tubules of the kidney, and sunitinib may protect against renal damage from DM. Sunitinib 14-23 vimentin Mus musculus 89-97 29652183-17 2018 The effect of sunitinib on experimental diabetic mice appears to be related to levels of vimentin, E-cadherin and S100 in the glomeruli and tubules of the kidney, and sunitinib may protect against renal damage from DM. Sunitinib 14-23 cadherin 1 Mus musculus 99-109 29652183-17 2018 The effect of sunitinib on experimental diabetic mice appears to be related to levels of vimentin, E-cadherin and S100 in the glomeruli and tubules of the kidney, and sunitinib may protect against renal damage from DM. Sunitinib 14-23 S100 calcium binding protein A1 Mus musculus 114-118 30636586-4 2018 We have presented that VEGF blockage in neoadjuvant setting via bevacizumab, aflibercept and sunitinib not only has revealed some promising benefits but also has shown a large negative outcome in the adjuvant trials. Sunitinib 93-102 vascular endothelial growth factor A Homo sapiens 23-27 30168120-2 2018 In 2005, with the advent of vascular endothelial growth factor (VEGF) tyrosine kinase inhibitor (TKI) therapy, the standard of care shifted to agents such as sunitinib and pazopanib. Sunitinib 158-167 vascular endothelial growth factor A Homo sapiens 28-62 30168120-2 2018 In 2005, with the advent of vascular endothelial growth factor (VEGF) tyrosine kinase inhibitor (TKI) therapy, the standard of care shifted to agents such as sunitinib and pazopanib. Sunitinib 158-167 vascular endothelial growth factor A Homo sapiens 64-68 30539696-8 2018 RESULTS: HepG2 cells with high CBS expression were less sensitive to DOX and sunitinib and knockdown of CBS significantly elevated the sensitivity to DOX and sunitinib in HepG2 cells. Sunitinib 77-86 cystathionine beta-synthase Homo sapiens 31-34 30539696-8 2018 RESULTS: HepG2 cells with high CBS expression were less sensitive to DOX and sunitinib and knockdown of CBS significantly elevated the sensitivity to DOX and sunitinib in HepG2 cells. Sunitinib 158-167 cystathionine beta-synthase Homo sapiens 104-107 30539696-9 2018 In contrast, CBS overexpression increased the resistance of DOX and sunitinib in BEL-7404 cells. Sunitinib 68-77 cystathionine beta-synthase Homo sapiens 13-16 29174910-0 2018 Corrigendum to "Enhanced expression of caveolin-1 possesses diagnostic and prognostic value and promotes cell migration, invasion and sunitinib resistance in the clear cell renal cell carcinoma" [Exp. Sunitinib 134-143 caveolin 1 Homo sapiens 39-49 28885866-5 2018 Of particular interest is the novel use of tyrosine kinase inhibitors (sunitinib and its derivatives) for the prevention and treatment of age-related ocular diseases via inhibition of the caspase-3, dual-leucine zipper kinase (DLK) and leucine zipper-bearing kinase (LZK) pathways. Sunitinib 71-80 caspase 3 Homo sapiens 188-197 28885866-5 2018 Of particular interest is the novel use of tyrosine kinase inhibitors (sunitinib and its derivatives) for the prevention and treatment of age-related ocular diseases via inhibition of the caspase-3, dual-leucine zipper kinase (DLK) and leucine zipper-bearing kinase (LZK) pathways. Sunitinib 71-80 mitogen-activated protein kinase kinase kinase 12 Homo sapiens 199-225 28885866-5 2018 Of particular interest is the novel use of tyrosine kinase inhibitors (sunitinib and its derivatives) for the prevention and treatment of age-related ocular diseases via inhibition of the caspase-3, dual-leucine zipper kinase (DLK) and leucine zipper-bearing kinase (LZK) pathways. Sunitinib 71-80 mitogen-activated protein kinase kinase kinase 12 Homo sapiens 227-230 28885866-5 2018 Of particular interest is the novel use of tyrosine kinase inhibitors (sunitinib and its derivatives) for the prevention and treatment of age-related ocular diseases via inhibition of the caspase-3, dual-leucine zipper kinase (DLK) and leucine zipper-bearing kinase (LZK) pathways. Sunitinib 71-80 mitogen-activated protein kinase kinase kinase 13 Homo sapiens 236-265 28885866-5 2018 Of particular interest is the novel use of tyrosine kinase inhibitors (sunitinib and its derivatives) for the prevention and treatment of age-related ocular diseases via inhibition of the caspase-3, dual-leucine zipper kinase (DLK) and leucine zipper-bearing kinase (LZK) pathways. Sunitinib 71-80 mitogen-activated protein kinase kinase kinase 13 Homo sapiens 267-270 29289530-9 2018 This model enables the study of anti-angiogenic drugs which target a specific factor/receptor pathway, as demonstrated by the use of the clinically approved sorafenib and sunitinib for targeting the VEGF-A/VEGFR-2 pathway. Sunitinib 171-180 vascular endothelial growth factor A Homo sapiens 199-205 29289530-9 2018 This model enables the study of anti-angiogenic drugs which target a specific factor/receptor pathway, as demonstrated by the use of the clinically approved sorafenib and sunitinib for targeting the VEGF-A/VEGFR-2 pathway. Sunitinib 171-180 kinase insert domain receptor Homo sapiens 206-213 30123861-6 2018 Flow cytometry showed that treatment with sunitinib, not sorafenib, significantly reduced the frequency of regulatory T cells (Tregs) and myeloid-derived suppressive cells (MDSCs) in tumor-bearing mice; and allowed splenic lymphocytes to produce equivalent levels of IFN-gamma and TNF-alpha in response to vaccination as that in wild type mice. Sunitinib 42-51 interferon gamma Mus musculus 267-276 30123861-6 2018 Flow cytometry showed that treatment with sunitinib, not sorafenib, significantly reduced the frequency of regulatory T cells (Tregs) and myeloid-derived suppressive cells (MDSCs) in tumor-bearing mice; and allowed splenic lymphocytes to produce equivalent levels of IFN-gamma and TNF-alpha in response to vaccination as that in wild type mice. Sunitinib 42-51 tumor necrosis factor Mus musculus 281-290 28966072-6 2018 PFS1 and 2 were defined as the time between the start of sunitinib and first progression and the time between dose-escalation and second progression respectively. Sunitinib 57-66 GINS complex subunit 2 Homo sapiens 0-10 30286478-10 2018 In 3 patients KIT mutations (e.g. pM541L, pV654A) known to be tissue-based biomarkers with level 1 evidence for the treatment with imatinib and sunitinib were found. Sunitinib 144-153 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 14-17 29290798-10 2018 SU inhibits signaling of vascular endothelial growth factor (VEGF), which is responsible for the sprouting of new vasculature, and GSI inhibits the Notch pathway, which is a key factor in the correct maturation of newly formed vasculature. Sunitinib 0-2 vascular endothelial growth factor A Mus musculus 25-59 29290798-10 2018 SU inhibits signaling of vascular endothelial growth factor (VEGF), which is responsible for the sprouting of new vasculature, and GSI inhibits the Notch pathway, which is a key factor in the correct maturation of newly formed vasculature. Sunitinib 0-2 vascular endothelial growth factor A Mus musculus 61-65 29290798-13 2018 Results The results show that MI can detect changes in VEGFR-2 expression in the group treated with SU within a week of the start of treatment, while differences in volume only become apparent after the mice have been treated for three weeks. Sunitinib 100-102 kinase insert domain protein receptor Mus musculus 55-62 28479237-16 2018 S-TRAC provides evidence that 1 year of adjuvant sunitinib in patients with higher risk locoregional disease increases the median time to recurrence. Sunitinib 49-58 T cell receptor alpha constant Homo sapiens 2-6 29399154-0 2018 Lysosome-associated membrane protein 2 (LAMP-2) expression induced by miR-194-5p downregulation contributes to sunitinib resistance in human renal cell carcinoma cells. Sunitinib 111-120 lysosomal associated membrane protein 2 Homo sapiens 40-46 29399154-0 2018 Lysosome-associated membrane protein 2 (LAMP-2) expression induced by miR-194-5p downregulation contributes to sunitinib resistance in human renal cell carcinoma cells. Sunitinib 111-120 membrane associated ring-CH-type finger 8 Homo sapiens 70-73 29399154-5 2018 Transfection of miR-194-5p, though not with negative control miR, in SR-ACHN cells could significantly inhibit cell proliferation following sunitinib treatment (2.5-40 microM; P<0.05). Sunitinib 140-149 membrane associated ring-CH-type finger 8 Homo sapiens 16-19 29399154-6 2018 Western blotting demonstrated that the expression of lysosome-associated membrane protein-2 (LAMP-2), which attenuates the anti-proliferative effect of sunitinib, was significantly higher in SR-ACHN than in ACHN cells (P<0.01). Sunitinib 152-161 lysosomal associated membrane protein 2 Homo sapiens 53-91 29399154-6 2018 Western blotting demonstrated that the expression of lysosome-associated membrane protein-2 (LAMP-2), which attenuates the anti-proliferative effect of sunitinib, was significantly higher in SR-ACHN than in ACHN cells (P<0.01). Sunitinib 152-161 lysosomal associated membrane protein 2 Homo sapiens 93-99 29399154-8 2018 These data suggested that miR-194-5p downregulation may be associated with sunitinib resistance via the induction of LAMP-2 expression in human RCC. Sunitinib 75-84 membrane associated ring-CH-type finger 8 Homo sapiens 26-29 29399154-8 2018 These data suggested that miR-194-5p downregulation may be associated with sunitinib resistance via the induction of LAMP-2 expression in human RCC. Sunitinib 75-84 lysosomal associated membrane protein 2 Homo sapiens 117-123 28986088-0 2018 Expression of tyrosine kinase receptor AXL is associated with worse outcome of metastatic renal cell carcinomas treated with sunitinib. Sunitinib 125-134 AXL receptor tyrosine kinase Homo sapiens 39-42 28986088-4 2018 Recently, others and our group suggested that the receptor tyrosine kinase AXL may modify the response to sunitinib. Sunitinib 106-115 AXL receptor tyrosine kinase Homo sapiens 75-78 28986088-5 2018 OBJECTIVE: To study the expression of AXL in a series patients with of mRCC treated with sunitinib and to correlate it with patient"s clinic-pathological features and therapeutic response. Sunitinib 89-98 AXL receptor tyrosine kinase Homo sapiens 38-41 28986088-10 2018 Moreover, AXL expression was associated with shorter progression to sunitinib. Sunitinib 68-77 AXL receptor tyrosine kinase Homo sapiens 10-13 28986088-12 2018 CONCLUSION: AXL expression was associated with worse clinical outcome and may be an important prognostic biomarker in sunitinib-treated patients with metastatic renal cell carcinoma. Sunitinib 118-127 AXL receptor tyrosine kinase Homo sapiens 12-15 29416656-4 2018 Western blot showed that propranolol and sunitinib combination significantly down-regulated phospho-Rb, phospho-ERK, Cyclin D1, and Cyclin E, but had no effect on Bax, Bcl-2, or cleaved PARP expression. Sunitinib 41-50 cyclin D1 Mus musculus 117-126 28978636-0 2017 EZH2 Modifies Sunitinib Resistance in Renal Cell Carcinoma by Kinome Reprogramming. Sunitinib 14-23 enhancer of zeste 2 polycomb repressive complex 2 subunit Homo sapiens 0-4 28978636-6 2017 Modulating EZH2 expression or activity suppressed phosphorylation of certain RTKs, restoring the antitumor effects of sunitinib in models of acquired or intrinsically resistant ccRCC. Sunitinib 118-127 enhancer of zeste 2 polycomb repressive complex 2 subunit Homo sapiens 11-15 28978636-7 2017 Overall, our results highlight EZH2 as a rational target for therapeutic intervention in sunitinib-resistant ccRCC as well as a predictive marker for RTKi response in this disease. Sunitinib 89-98 enhancer of zeste 2 polycomb repressive complex 2 subunit Homo sapiens 31-35 30381648-11 2018 As a result, the ICER associated with replacing IFN-alpha with sunitinib was 22,695,839 yen/QALYs. Sunitinib 63-72 interferon alpha 1 Homo sapiens 48-57 28745317-8 2017 Moreover, we demonstrated that RCN2 knockout sensitized HCC cells to tyrosine kinase inhibitors, including erlotinib, lapatinib and sunitinib. Sunitinib 132-141 reticulocalbin 2 Homo sapiens 31-35 29416656-4 2018 Western blot showed that propranolol and sunitinib combination significantly down-regulated phospho-Rb, phospho-ERK, Cyclin D1, and Cyclin E, but had no effect on Bax, Bcl-2, or cleaved PARP expression. Sunitinib 41-50 B cell leukemia/lymphoma 2 Mus musculus 168-173 29416656-4 2018 Western blot showed that propranolol and sunitinib combination significantly down-regulated phospho-Rb, phospho-ERK, Cyclin D1, and Cyclin E, but had no effect on Bax, Bcl-2, or cleaved PARP expression. Sunitinib 41-50 mitogen-activated protein kinase 1 Mus musculus 112-115 29416656-4 2018 Western blot showed that propranolol and sunitinib combination significantly down-regulated phospho-Rb, phospho-ERK, Cyclin D1, and Cyclin E, but had no effect on Bax, Bcl-2, or cleaved PARP expression. Sunitinib 41-50 poly (ADP-ribose) polymerase family, member 1 Mus musculus 186-190 29416656-7 2018 This synergistic effect between propranolol and sunitinib to inhibit MM proliferation was through suppressing ERK/Cyclin D1/Rb/Cyclin E pathways and inducing G0/G1/S phase arrest, rather than by inducing tumor cell apoptosis. Sunitinib 48-57 mitogen-activated protein kinase 1 Mus musculus 110-113 29416656-7 2018 This synergistic effect between propranolol and sunitinib to inhibit MM proliferation was through suppressing ERK/Cyclin D1/Rb/Cyclin E pathways and inducing G0/G1/S phase arrest, rather than by inducing tumor cell apoptosis. Sunitinib 48-57 cyclin D1 Mus musculus 114-123 28898759-8 2017 In addition, CONPs downregulate the expression of AXL, MET, AKT, and ERK to recover sunitinib responsiveness in RCC cells with sunitinib resistance (SR) and may therefore facilitate the development of promising new pathways to treat patients with acquired SRRCC. Sunitinib 84-93 AXL receptor tyrosine kinase Homo sapiens 50-53 29290991-7 2017 In addition, we identified adrenomedullin (ADM), previously shown to be implicated in sunitinib resistance, as the earliest response gene upon ALK inhibition. Sunitinib 86-95 ALK receptor tyrosine kinase Homo sapiens 143-146 28811127-2 2017 Using rat Langendorff heart model and human acute myeloid leukaemia 60 (HL60) cell line we detected the involvement of protein kinase C (PKC) alpha during Sunitinib-induced cardiotoxicity and the effect of Sunitinib on cancer progression. Sunitinib 155-164 protein kinase C alpha Homo sapiens 119-147 28811127-12 2017 This study reveals that A3 adenosine receptor activation by IB-MECA attenuates Sunitinib-induced cardiotoxicity through the involvement of PKCalpha. Sunitinib 79-88 protein kinase C alpha Homo sapiens 139-147 28898759-8 2017 In addition, CONPs downregulate the expression of AXL, MET, AKT, and ERK to recover sunitinib responsiveness in RCC cells with sunitinib resistance (SR) and may therefore facilitate the development of promising new pathways to treat patients with acquired SRRCC. Sunitinib 84-93 AKT serine/threonine kinase 1 Homo sapiens 60-63 28729403-10 2017 Sunitinib also induced lysosomal membrane permeabilization, which further increased in the presence of chloroquine or knockdown of lysosome-associated membrane protein (LAMP2). Sunitinib 0-9 lysosomal associated membrane protein 2 Homo sapiens 169-174 28898759-8 2017 In addition, CONPs downregulate the expression of AXL, MET, AKT, and ERK to recover sunitinib responsiveness in RCC cells with sunitinib resistance (SR) and may therefore facilitate the development of promising new pathways to treat patients with acquired SRRCC. Sunitinib 84-93 mitogen-activated protein kinase 1 Homo sapiens 69-72 28898759-8 2017 In addition, CONPs downregulate the expression of AXL, MET, AKT, and ERK to recover sunitinib responsiveness in RCC cells with sunitinib resistance (SR) and may therefore facilitate the development of promising new pathways to treat patients with acquired SRRCC. Sunitinib 127-136 AXL receptor tyrosine kinase Homo sapiens 50-53 28898759-8 2017 In addition, CONPs downregulate the expression of AXL, MET, AKT, and ERK to recover sunitinib responsiveness in RCC cells with sunitinib resistance (SR) and may therefore facilitate the development of promising new pathways to treat patients with acquired SRRCC. Sunitinib 127-136 mitogen-activated protein kinase 1 Homo sapiens 69-72 29262588-2 2017 Agents targeting angiogenesis and mTOR, such as sunitinib and everolimus (RAD001), have been shown to result in progression-free survival of approximately 11 months in patients with advanced pNETs. Sunitinib 48-57 mechanistic target of rapamycin kinase Homo sapiens 34-38 29187872-0 2017 Tumoral ANXA1 Is a Predictive Marker for Sunitinib Treatment of Renal Cancer Patients. Sunitinib 41-50 annexin A1 Homo sapiens 8-13 29187872-4 2017 Our aim with the current study was to assess annexin A1 (ANXA1), which stimulates angiogenesis, as a predictive marker for sunitinib therapy in mRCC patients. Sunitinib 123-132 annexin A1 Homo sapiens 45-55 29187872-4 2017 Our aim with the current study was to assess annexin A1 (ANXA1), which stimulates angiogenesis, as a predictive marker for sunitinib therapy in mRCC patients. Sunitinib 123-132 annexin A1 Homo sapiens 57-62 29187872-13 2017 Conclusions: Our results indicate that cytoplasmic expression of ANXA1 is a negative predictive marker for sunitinib therapy in mRCC patients. Sunitinib 107-116 annexin A1 Homo sapiens 65-70 29212269-0 2017 Low neighbor of Brca1 gene expression predicts poor clinical outcome and resistance of sunitinib in clear cell renal cell carcinoma. Sunitinib 87-96 BRCA1 DNA repair associated Homo sapiens 16-21 29212269-8 2017 Low level of NBR1 mRNA showed a significance poor prognostic of overall survival (OS), disease-free survival (DFS) with univariate and multivariate analyses in ccRCC patients and sunitinib resistance. Sunitinib 179-188 NBR1 autophagy cargo receptor Homo sapiens 13-17 29212269-9 2017 Conclusions: Taken together, our results suggest that low level of NBR1 can predict poor clinical outcome and resistance of sunitinib in patients with ccRCC. Sunitinib 124-133 NBR1 autophagy cargo receptor Homo sapiens 67-71 28805145-7 2017 An increase in extracellular TGase activity on the HPT cell membrane was observed in a dose-dependent manner after treatment with Sunitinib malate. Sunitinib 130-146 transglutaminase 1 Homo sapiens 29-34 28850564-0 2017 CXCL7 is a predictive marker of sunitinib efficacy in clear cell renal cell carcinomas. Sunitinib 32-41 pro-platelet basic protein Homo sapiens 0-5 28330808-1 2017 BACKGROUND: Sunitinib, the vascular endothelial growth factor pathway inhibitor, is an established standard-of-care for advanced renal cell carcinoma (RCC). Sunitinib 12-21 vascular endothelial growth factor A Homo sapiens 27-61 28850564-9 2017 CONCLUSIONS: CXCL7 may be considered as a predictive marker of sunitinib efficacy for ccRCC patients. Sunitinib 63-72 pro-platelet basic protein Homo sapiens 13-18 29296523-8 2017 Mechanistically, sunitinib treatment primes the antitumor immune response by significantly decreasing Treg frequency, reducing TGF-beta and IL-10 production by Tregs, and also protecting TAS CD8+ T cells from tumor-induced deletion in the setting of HCC. Sunitinib 17-26 transforming growth factor, beta 1 Mus musculus 127-135 28756150-7 2017 The cytotoxicity of 4-CF was reversed by co-treatment with the VEGF and Akt inhibitors sunitinib and perifosine, respectively or by the addition of neutralizing VEGF antibodies. Sunitinib 87-96 vascular endothelial growth factor A Mus musculus 63-67 28756150-7 2017 The cytotoxicity of 4-CF was reversed by co-treatment with the VEGF and Akt inhibitors sunitinib and perifosine, respectively or by the addition of neutralizing VEGF antibodies. Sunitinib 87-96 thymoma viral proto-oncogene 1 Mus musculus 72-75 28993730-8 2017 However, targeting the altered biological pathways (mTOR, PDGFRB, FGF2, HDAC) guided identification of possibly beneficial treatment with a combination of sirolimus, thalidomide, sunitinib, and vorinostat. Sunitinib 179-188 mechanistic target of rapamycin kinase Homo sapiens 52-56 28993730-8 2017 However, targeting the altered biological pathways (mTOR, PDGFRB, FGF2, HDAC) guided identification of possibly beneficial treatment with a combination of sirolimus, thalidomide, sunitinib, and vorinostat. Sunitinib 179-188 histone deacetylase 9 Homo sapiens 72-76 29296523-8 2017 Mechanistically, sunitinib treatment primes the antitumor immune response by significantly decreasing Treg frequency, reducing TGF-beta and IL-10 production by Tregs, and also protecting TAS CD8+ T cells from tumor-induced deletion in the setting of HCC. Sunitinib 17-26 interleukin 10 Mus musculus 140-145 29033582-0 2017 HIF pathway and c-Myc as biomarkers for response to sunitinib in metastatic clear-cell renal cell carcinoma. Sunitinib 52-61 MYC proto-oncogene, bHLH transcription factor Homo sapiens 16-21 28870911-5 2017 Apoptotic death induced by sunitinib in MCF7 cells was mediated by activation of caspase-3 and p53 mRNA and protein expression and an increase in the percentage of apoptotic cells (40%) as determined by flow cytometry. Sunitinib 27-36 caspase 3 Homo sapiens 81-90 28684115-0 2017 Enhanced expression of caveolin-1 possesses diagnostic and prognostic value and promotes cell migration, invasion and sunitinib resistance in the clear cell renal cell carcinoma. Sunitinib 118-127 caveolin 1 Homo sapiens 23-33 28684115-6 2017 Moreover, CAV1 expression was up-regulated in sunitinib-resistant renal cancer cell lines, and its overexpression promoted sunitinib resistance. Sunitinib 46-55 caveolin 1 Homo sapiens 10-14 28684115-6 2017 Moreover, CAV1 expression was up-regulated in sunitinib-resistant renal cancer cell lines, and its overexpression promoted sunitinib resistance. Sunitinib 123-132 caveolin 1 Homo sapiens 10-14 28684115-7 2017 In general, our results confirm that CAV1 plays an important role in the metastasis of kidney cancer and induces sunitinib resistance, so CAV1 function suppression may become a promising clinical treatment strategy during renal cell carcinoma metastasis and sunitinib resistance. Sunitinib 113-122 caveolin 1 Homo sapiens 37-41 28684115-7 2017 In general, our results confirm that CAV1 plays an important role in the metastasis of kidney cancer and induces sunitinib resistance, so CAV1 function suppression may become a promising clinical treatment strategy during renal cell carcinoma metastasis and sunitinib resistance. Sunitinib 258-267 caveolin 1 Homo sapiens 37-41 28684115-7 2017 In general, our results confirm that CAV1 plays an important role in the metastasis of kidney cancer and induces sunitinib resistance, so CAV1 function suppression may become a promising clinical treatment strategy during renal cell carcinoma metastasis and sunitinib resistance. Sunitinib 258-267 caveolin 1 Homo sapiens 138-142 29262547-10 2017 In addition, crotonoside alone or the combination of sunitinib/RFP966/HPOB exhibited a significant post-inhibition effect in AML cells by the inhibition of FLT3 and HDAC3/6. Sunitinib 53-62 fms related receptor tyrosine kinase 3 Homo sapiens 156-160 29262547-10 2017 In addition, crotonoside alone or the combination of sunitinib/RFP966/HPOB exhibited a significant post-inhibition effect in AML cells by the inhibition of FLT3 and HDAC3/6. Sunitinib 53-62 histone deacetylase 3 Homo sapiens 165-170 28870911-5 2017 Apoptotic death induced by sunitinib in MCF7 cells was mediated by activation of caspase-3 and p53 mRNA and protein expression and an increase in the percentage of apoptotic cells (40%) as determined by flow cytometry. Sunitinib 27-36 tumor protein p53 Homo sapiens 95-98 28870911-7 2017 Mechanistically, blocking of de novo RNA synthesis by actinomycin D significantly inhibited sunitinib-induced expression of p53 mRNA, but not that of caspase-3, indicating involvement of a transcriptional mechanism. Sunitinib 92-101 tumor protein p53 Homo sapiens 124-127 28870911-9 2017 In addition, sunitinib exhibited concentration-dependent induction of oxidative stress genes (heme oxygenase 1 and glutathione transferase A1) through the nuclear factor erythroid 2-related factor 2 pathway. Sunitinib 13-22 heme oxygenase 1 Homo sapiens 94-110 28870911-9 2017 In addition, sunitinib exhibited concentration-dependent induction of oxidative stress genes (heme oxygenase 1 and glutathione transferase A1) through the nuclear factor erythroid 2-related factor 2 pathway. Sunitinib 13-22 NFE2 like bZIP transcription factor 2 Homo sapiens 155-198 28796254-0 2017 Sunitinib induces genomic instability of renal carcinoma cells through affecting the interaction of LC3-II and PARP-1. Sunitinib 0-9 microtubule associated protein 1 light chain 3 alpha Homo sapiens 100-103 28778562-5 2017 Compound 2 exhibited angiogenesis effect against sunitinib-induced damage on intersegmental blood vessels in Tg (flk1: EGFP) and Tg (fli1: nEGFP) transgenic zebrafish at concentrations of 3.13, 6.25, 12.50, and 25.00muM. Sunitinib 49-58 kinase insert domain receptor (a type III receptor tyrosine kinase) Danio rerio 113-117 28778562-5 2017 Compound 2 exhibited angiogenesis effect against sunitinib-induced damage on intersegmental blood vessels in Tg (flk1: EGFP) and Tg (fli1: nEGFP) transgenic zebrafish at concentrations of 3.13, 6.25, 12.50, and 25.00muM. Sunitinib 49-58 Fli-1 proto-oncogene, ETS transcription factor Danio rerio 133-137 28859644-0 2017 Predictive value of C-reactive protein in patients treated with sunitinib for metastatic clear cell renal cell carcinoma. Sunitinib 64-73 C-reactive protein Homo sapiens 20-38 28859644-13 2017 CONCLUSION: Baseline CRP was a significant predictive factor of sunitinib response and a prognostic factor of survival. Sunitinib 64-73 C-reactive protein Homo sapiens 21-24 28624441-0 2017 Hsp90 inhibitor geldanamycin attenuates the cytotoxicity of sunitinib in cardiomyocytes via inhibition of the autophagy pathway. Sunitinib 60-69 heat shock protein 90 alpha family class A member 1 Homo sapiens 0-5 28624441-3 2017 Here, we examined the effect of geldanamycin, an inhibitor of heat shock protein (Hsp) 90, on sunitinib-induced cytotoxicity in cardiomyocytes. Sunitinib 94-103 heat shock protein 90 alpha family class A member 1 Homo sapiens 62-89 28624441-4 2017 First, we found that treatment with geldanamycin or other Hsp90 inhibitors (tanespimycin, ganetespib, or BIIB021) significantly attenuated sunitinib-induced cytotoxicity in rat H9c2 cardiomyocytes, suggesting a drug-class effect of Hsp90 inhibitors. Sunitinib 139-148 heat shock protein 90 alpha family class A member 1 Rattus norvegicus 58-63 28624441-4 2017 First, we found that treatment with geldanamycin or other Hsp90 inhibitors (tanespimycin, ganetespib, or BIIB021) significantly attenuated sunitinib-induced cytotoxicity in rat H9c2 cardiomyocytes, suggesting a drug-class effect of Hsp90 inhibitors. Sunitinib 139-148 heat shock protein 90 alpha family class A member 1 Rattus norvegicus 232-237 28624441-5 2017 We then examined the mechanisms underlying sunitinib-induced cytotoxicity and found that sunitinib induced autophagy in H9c2 cells and that pretreatment with geldanamycin inhibited the induction of autophagy by promoting degradation of the autophagy-related proteins Atg7, Beclin-1, and ULK1. Sunitinib 43-52 autophagy related 7 Rattus norvegicus 267-271 28624441-5 2017 We then examined the mechanisms underlying sunitinib-induced cytotoxicity and found that sunitinib induced autophagy in H9c2 cells and that pretreatment with geldanamycin inhibited the induction of autophagy by promoting degradation of the autophagy-related proteins Atg7, Beclin-1, and ULK1. Sunitinib 43-52 beclin 1 Rattus norvegicus 273-281 28624441-5 2017 We then examined the mechanisms underlying sunitinib-induced cytotoxicity and found that sunitinib induced autophagy in H9c2 cells and that pretreatment with geldanamycin inhibited the induction of autophagy by promoting degradation of the autophagy-related proteins Atg7, Beclin-1, and ULK1. Sunitinib 43-52 unc-51 like autophagy activating kinase 1 Rattus norvegicus 287-291 28624441-10 2017 Thus, the further investigation of combination or sequential treatment with an Hsp90 inhibitor and sunitinib is warranted as a potential strategy of attenuating the cardiotoxicity associated with sunitinib administration in the clinical setting. Sunitinib 196-205 heat shock protein 90 alpha family class A member 1 Homo sapiens 79-84 28796254-0 2017 Sunitinib induces genomic instability of renal carcinoma cells through affecting the interaction of LC3-II and PARP-1. Sunitinib 0-9 poly(ADP-ribose) polymerase 1 Homo sapiens 111-117 28978162-0 2017 Effects of VEGF and VEGFR polymorphisms on the outcome of patients with metastatic renal cell carcinoma treated with sunitinib: a systematic review and meta-analysis. Sunitinib 117-126 kinase insert domain receptor Homo sapiens 20-25 28978162-4 2017 VEGFR1 rs9582036 AA/AC carriers and rs9554320 CC/AC carriers had more favorable overall survival (OS) in patients with mRCC treated with sunitinib (n = 3), but not in progression-free survival (PFS). Sunitinib 137-146 fms related receptor tyrosine kinase 1 Homo sapiens 0-6 28978162-7 2017 VEGFR2 rs1870377 was verified to be associated with sunitinib OS (n = 1). Sunitinib 52-61 kinase insert domain receptor Homo sapiens 0-6 28978162-8 2017 Furthermore, patients with VEGFR3 rs307826 and rs307821 had shorter PFS and OS during sunitinib therapy (n = 2, respectively). Sunitinib 86-95 fms related receptor tyrosine kinase 4 Homo sapiens 27-33 28978162-9 2017 Our results suggested that VEGF and VEGFR polymorphisms were associated with outcomes in sunitinib treated mRCC patients, especially VEGFR1 polymorphisms. Sunitinib 89-98 vascular endothelial growth factor A Homo sapiens 27-31 28978162-9 2017 Our results suggested that VEGF and VEGFR polymorphisms were associated with outcomes in sunitinib treated mRCC patients, especially VEGFR1 polymorphisms. Sunitinib 89-98 kinase insert domain receptor Homo sapiens 36-41 28978162-9 2017 Our results suggested that VEGF and VEGFR polymorphisms were associated with outcomes in sunitinib treated mRCC patients, especially VEGFR1 polymorphisms. Sunitinib 89-98 fms related receptor tyrosine kinase 1 Homo sapiens 133-139 28544544-4 2017 Screening of a library of compounds containing approximately 90 molecular-targeting drugs revealed that this process was suppressed by the insulin-like growth factor-1 (IGF-1) receptor (IGF-1R)-specific kinase inhibitor OSI-906, as well as the multikinase inhibitors axitinib and sunitinib. Sunitinib 280-289 insulin like growth factor 1 receptor Homo sapiens 186-192 28449948-3 2017 This study aims to analyse whether sunitinib has specific and direct effects on insulin secreting beta-cells. Sunitinib 35-44 insulin Homo sapiens 80-87 28449948-8 2017 Sunitinib further augmented insulin secretion in the presence of elevated cAMP levels and the FFAR1 agonists. Sunitinib 0-9 free fatty acid receptor 1 Homo sapiens 94-99 28819401-10 2017 Analyses of two open source, RNA expression data sets on sunitinib response revealed that SLC10A2 was downregulated in tyrosine kinase inhibitor-resistant samples. Sunitinib 57-66 solute carrier family 10 member 2 Homo sapiens 90-97 28237182-0 2017 Correlation of c-Met Expression and Outcome in Patients With Renal Cell Carcinoma Treated With Sunitinib. Sunitinib 95-104 MET proto-oncogene, receptor tyrosine kinase Homo sapiens 15-20 28237182-3 2017 The main objective was to evaluate c-Met expression in sunitinib-treated patients with mRCC, including patients with bone metastases. Sunitinib 55-64 MET proto-oncogene, receptor tyrosine kinase Homo sapiens 35-40 28237182-8 2017 CONCLUSIONS: High c-Met expression was associated with poor survival in patients with mRCC treated with sunitinib. Sunitinib 104-113 MET proto-oncogene, receptor tyrosine kinase Homo sapiens 18-23 28672196-2 2017 Therapeutic alternatives such as other tyrosine kinase inhibitors, VEGF inhibitors, or mTOR inhibitors emphasize the clinical need to predict the patient"s response to sunitinib therapy before treatment initiation. Sunitinib 168-177 vascular endothelial growth factor A Homo sapiens 67-71 28672196-2 2017 Therapeutic alternatives such as other tyrosine kinase inhibitors, VEGF inhibitors, or mTOR inhibitors emphasize the clinical need to predict the patient"s response to sunitinib therapy before treatment initiation. Sunitinib 168-177 mechanistic target of rapamycin kinase Homo sapiens 87-91 28903416-7 2017 786-O RCC cells secrete high IL-6 levels after low dose stimulation with the TKIs sorafenib, sunitinib and pazopanib, inducing activation of AKT-mTOR pathway, NFkappaB, HIF-2alpha and VEGF expression. Sunitinib 93-102 interleukin 6 Homo sapiens 29-33 28903416-7 2017 786-O RCC cells secrete high IL-6 levels after low dose stimulation with the TKIs sorafenib, sunitinib and pazopanib, inducing activation of AKT-mTOR pathway, NFkappaB, HIF-2alpha and VEGF expression. Sunitinib 93-102 AKT serine/threonine kinase 1 Homo sapiens 141-144 28903416-7 2017 786-O RCC cells secrete high IL-6 levels after low dose stimulation with the TKIs sorafenib, sunitinib and pazopanib, inducing activation of AKT-mTOR pathway, NFkappaB, HIF-2alpha and VEGF expression. Sunitinib 93-102 mechanistic target of rapamycin kinase Homo sapiens 145-149 28903416-7 2017 786-O RCC cells secrete high IL-6 levels after low dose stimulation with the TKIs sorafenib, sunitinib and pazopanib, inducing activation of AKT-mTOR pathway, NFkappaB, HIF-2alpha and VEGF expression. Sunitinib 93-102 endothelial PAS domain protein 1 Homo sapiens 169-179 28903416-7 2017 786-O RCC cells secrete high IL-6 levels after low dose stimulation with the TKIs sorafenib, sunitinib and pazopanib, inducing activation of AKT-mTOR pathway, NFkappaB, HIF-2alpha and VEGF expression. Sunitinib 93-102 vascular endothelial growth factor A Homo sapiens 184-188 28196874-10 2017 These elevations similarly tended to occur early and resolved over time.Conclusions: On average, patients with mRCC receiving sunitinib exhibit modest declines in LVEF and nonsignificant changes in hsTnI and BNP. Sunitinib 126-135 natriuretic peptide B Homo sapiens 208-211 29086859-2 2017 Compared to sunitinib, the C(5)-Br derivative of 2-pyrrolidone-fused (2-oxoindolin-3-ylidene)methylpyrrole has significantly greater in vitro activities against VEGFR-2, PDGFRbeta, and tube formation. Sunitinib 12-21 kinase insert domain receptor Homo sapiens 161-168 29086859-8 2017 Graphical abstract IC50 comparison of sunitinib, 14, and 15 against VEGFR-2 and PDGFRbeta. Sunitinib 38-47 platelet derived growth factor receptor beta Homo sapiens 80-89 28796048-10 2017 Four patients (33.3%) developed disease progression during imatinib treatment after initial resection, but all of these patients regained disease control when the treatment was altered to sunitinib targeted therapy.SDH-deficient GISTs arise exclusively in the stomach and account for approximately 7.4% (12/162) of gastric GISTs. Sunitinib 188-197 succinate dehydrogenase complex iron sulfur subunit B Homo sapiens 215-218 28550387-0 2017 Correlation of c-MET Expression with PD-L1 Expression in Metastatic Clear Cell Renal Cell Carcinoma Treated by Sunitinib First-Line Therapy. Sunitinib 111-120 MET proto-oncogene, receptor tyrosine kinase Homo sapiens 15-20 28550387-0 2017 Correlation of c-MET Expression with PD-L1 Expression in Metastatic Clear Cell Renal Cell Carcinoma Treated by Sunitinib First-Line Therapy. Sunitinib 111-120 CD274 molecule Homo sapiens 37-42 28572533-7 2017 We found that the anti-angiogenic TKI sunitinib disrupted the balance between HIF-1alpha and HIF-2alpha in RCCs and led to a protective effect on HUVECs against sunitinib treatment when cultured with conditioned medium. Sunitinib 38-47 hypoxia inducible factor 1 subunit alpha Homo sapiens 78-88 28572533-7 2017 We found that the anti-angiogenic TKI sunitinib disrupted the balance between HIF-1alpha and HIF-2alpha in RCCs and led to a protective effect on HUVECs against sunitinib treatment when cultured with conditioned medium. Sunitinib 38-47 endothelial PAS domain protein 1 Homo sapiens 93-103 28572533-8 2017 Mechanistically, RCCs treated with sunitinib resulted in down-regulation of HIF-1alpha, but not HIF-2alpha, through reduction of both mRNA and protein levels. Sunitinib 35-44 hypoxia inducible factor 1 subunit alpha Homo sapiens 76-86 28572533-8 2017 Mechanistically, RCCs treated with sunitinib resulted in down-regulation of HIF-1alpha, but not HIF-2alpha, through reduction of both mRNA and protein levels. Sunitinib 35-44 endothelial PAS domain protein 1 Homo sapiens 96-106 28572533-9 2017 The down-regulation of HIF-1alpha by sunitinib occurred via hypoxia associated factor (HAF), which also enhanced HIF-2alpha transactivation activity to increase the production of pro-angiogenic factors and cytokines and promote HUVEC proliferation. Sunitinib 37-46 hypoxia inducible factor 1 subunit alpha Homo sapiens 23-33 28572533-9 2017 The down-regulation of HIF-1alpha by sunitinib occurred via hypoxia associated factor (HAF), which also enhanced HIF-2alpha transactivation activity to increase the production of pro-angiogenic factors and cytokines and promote HUVEC proliferation. Sunitinib 37-46 coagulation factor XII Homo sapiens 60-85 28572533-9 2017 The down-regulation of HIF-1alpha by sunitinib occurred via hypoxia associated factor (HAF), which also enhanced HIF-2alpha transactivation activity to increase the production of pro-angiogenic factors and cytokines and promote HUVEC proliferation. Sunitinib 37-46 coagulation factor XII Homo sapiens 87-90 28572533-9 2017 The down-regulation of HIF-1alpha by sunitinib occurred via hypoxia associated factor (HAF), which also enhanced HIF-2alpha transactivation activity to increase the production of pro-angiogenic factors and cytokines and promote HUVEC proliferation. Sunitinib 37-46 endothelial PAS domain protein 1 Homo sapiens 113-123 28978115-1 2017 OBJECTIVE: This study aims to investigate biological behavior changes in a murine lung cancer cell characterized by acquired resistance to sunitinib, a potent inhibitor of multiple-targeted receptor tyrosine kinase. Sunitinib 139-148 TYRO3 protein tyrosine kinase 3 Mus musculus 190-214 28781191-11 2017 The increased anti-factor Xa levels during combination treatment suggest that sunitinib might increase the anticoagulation activity of dalteparin. Sunitinib 78-87 coagulation factor X Homo sapiens 19-28 28640223-3 2017 The TKI erlotinib targets the epidermal growth factor receptor (EGFR), whereas sunitinib targets primarily vascular endothelial growth factor receptor (VEGFR) and platelet-derived growth factor receptor (PDGFR).TKIs that impact the function of non-malignant cells and have on- and off-target toxicities, including cardiotoxicities. Sunitinib 79-88 platelet derived growth factor receptor, beta polypeptide Mus musculus 163-202 29662544-6 2017 Sunitinib has also been tested in the adjuvant setting, within the ASSURE and S-TRAC trials, with opposite results; indeed, equivocal risk-stratification criteria, as well as immature overall survival (OS) data prevent any definitive conclusion on this important issue. Sunitinib 0-9 T cell receptor alpha constant Homo sapiens 80-84 28287633-10 2017 On the other hand, the VASH1 density was significantly higher in the metastatic ccRCCs treated with sunitinib compared with non-treated ones (P=0.010), indicating that VASH1 may be associated with the resistance of ECs to sunitinib treatment. Sunitinib 100-109 vasohibin 1 Homo sapiens 23-28 28287633-10 2017 On the other hand, the VASH1 density was significantly higher in the metastatic ccRCCs treated with sunitinib compared with non-treated ones (P=0.010), indicating that VASH1 may be associated with the resistance of ECs to sunitinib treatment. Sunitinib 100-109 vasohibin 1 Homo sapiens 168-173 28287633-10 2017 On the other hand, the VASH1 density was significantly higher in the metastatic ccRCCs treated with sunitinib compared with non-treated ones (P=0.010), indicating that VASH1 may be associated with the resistance of ECs to sunitinib treatment. Sunitinib 222-231 vasohibin 1 Homo sapiens 168-173 28693255-1 2017 Sunitinib (SU) is a small molecule that inhibits the receptor tyrosine kinase (RTK) signaling pathway, and has been clinically used to treat advanced renal cell carcinoma (RCC). Sunitinib 0-9 ret proto-oncogene Homo sapiens 53-77 28693255-1 2017 Sunitinib (SU) is a small molecule that inhibits the receptor tyrosine kinase (RTK) signaling pathway, and has been clinically used to treat advanced renal cell carcinoma (RCC). Sunitinib 0-9 ret proto-oncogene Homo sapiens 79-82 28693255-1 2017 Sunitinib (SU) is a small molecule that inhibits the receptor tyrosine kinase (RTK) signaling pathway, and has been clinically used to treat advanced renal cell carcinoma (RCC). Sunitinib 11-13 ret proto-oncogene Homo sapiens 53-77 28693255-1 2017 Sunitinib (SU) is a small molecule that inhibits the receptor tyrosine kinase (RTK) signaling pathway, and has been clinically used to treat advanced renal cell carcinoma (RCC). Sunitinib 11-13 ret proto-oncogene Homo sapiens 79-82 28640223-3 2017 The TKI erlotinib targets the epidermal growth factor receptor (EGFR), whereas sunitinib targets primarily vascular endothelial growth factor receptor (VEGFR) and platelet-derived growth factor receptor (PDGFR).TKIs that impact the function of non-malignant cells and have on- and off-target toxicities, including cardiotoxicities. Sunitinib 79-88 platelet derived growth factor receptor, beta polypeptide Mus musculus 204-209 28376387-0 2017 Comprehensive insight into the binding of sunitinib, a multi-targeted anticancer drug to human serum albumin. Sunitinib 42-51 albumin Homo sapiens 95-108 28376387-1 2017 Binding studies between a multi-targeted anticancer drug, sunitinib (SU) and human serum albumin (HSA) were made using fluorescence, UV-vis absorption, circular dichroism (CD) and molecular docking analysis. Sunitinib 58-67 albumin Homo sapiens 83-96 28376387-1 2017 Binding studies between a multi-targeted anticancer drug, sunitinib (SU) and human serum albumin (HSA) were made using fluorescence, UV-vis absorption, circular dichroism (CD) and molecular docking analysis. Sunitinib 69-71 albumin Homo sapiens 83-96 28327411-0 2017 Gas6-Axl signaling in presence of Sunitinib is enhanced, diversified and sustained in renal tumor cells, resulting in tumor-progressive advantages. Sunitinib 34-43 AXL receptor tyrosine kinase Homo sapiens 5-8 28422745-2 2017 We rationally designed and evaluated preclinically a novel sunitinib analogue, SAP, with favourable pharmacological properties and the ability to be readily conjugated to a targeting peptide or antibody for active tumour targeting.SAP was evaluated in silico and in vitro in order to verify target engagement (e.g., VEGFR2). Sunitinib 59-68 SH2 domain containing 1A Mus musculus 79-82 28422745-2 2017 We rationally designed and evaluated preclinically a novel sunitinib analogue, SAP, with favourable pharmacological properties and the ability to be readily conjugated to a targeting peptide or antibody for active tumour targeting.SAP was evaluated in silico and in vitro in order to verify target engagement (e.g., VEGFR2). Sunitinib 59-68 SH2 domain containing 1A Mus musculus 231-234 28422745-2 2017 We rationally designed and evaluated preclinically a novel sunitinib analogue, SAP, with favourable pharmacological properties and the ability to be readily conjugated to a targeting peptide or antibody for active tumour targeting.SAP was evaluated in silico and in vitro in order to verify target engagement (e.g., VEGFR2). Sunitinib 59-68 kinase insert domain protein receptor Mus musculus 316-322 28327411-0 2017 Gas6-Axl signaling in presence of Sunitinib is enhanced, diversified and sustained in renal tumor cells, resulting in tumor-progressive advantages. Sunitinib 34-43 growth arrest specific 6 Homo sapiens 0-4 28327411-2 2017 Investigation of Gas6-mediated Axl signaling in CCRCC and endothelial cells reveals a Sunitinib resistant Gas6-Axl signaling that is sustained and enhanced and specifically triggers downstream AKT and PRAS40 activation in an intensified manner. Sunitinib 86-95 growth arrest specific 6 Homo sapiens 17-21 28327411-2 2017 Investigation of Gas6-mediated Axl signaling in CCRCC and endothelial cells reveals a Sunitinib resistant Gas6-Axl signaling that is sustained and enhanced and specifically triggers downstream AKT and PRAS40 activation in an intensified manner. Sunitinib 86-95 AXL receptor tyrosine kinase Homo sapiens 31-34 28327411-2 2017 Investigation of Gas6-mediated Axl signaling in CCRCC and endothelial cells reveals a Sunitinib resistant Gas6-Axl signaling that is sustained and enhanced and specifically triggers downstream AKT and PRAS40 activation in an intensified manner. Sunitinib 86-95 growth arrest specific 6 Homo sapiens 106-110 28327411-2 2017 Investigation of Gas6-mediated Axl signaling in CCRCC and endothelial cells reveals a Sunitinib resistant Gas6-Axl signaling that is sustained and enhanced and specifically triggers downstream AKT and PRAS40 activation in an intensified manner. Sunitinib 86-95 AXL receptor tyrosine kinase Homo sapiens 111-114 28327411-2 2017 Investigation of Gas6-mediated Axl signaling in CCRCC and endothelial cells reveals a Sunitinib resistant Gas6-Axl signaling that is sustained and enhanced and specifically triggers downstream AKT and PRAS40 activation in an intensified manner. Sunitinib 86-95 AKT serine/threonine kinase 1 Homo sapiens 193-196 28327411-2 2017 Investigation of Gas6-mediated Axl signaling in CCRCC and endothelial cells reveals a Sunitinib resistant Gas6-Axl signaling that is sustained and enhanced and specifically triggers downstream AKT and PRAS40 activation in an intensified manner. Sunitinib 86-95 AKT1 substrate 1 Homo sapiens 201-207 28327411-3 2017 Gas6-induced Axl signaling in presence of Sunitinib is also diversified displaying onset of Axl-dependent EGFR and METR activation and activation of classical MAPK pathways. Sunitinib 42-51 growth arrest specific 6 Homo sapiens 0-4 28327411-3 2017 Gas6-induced Axl signaling in presence of Sunitinib is also diversified displaying onset of Axl-dependent EGFR and METR activation and activation of classical MAPK pathways. Sunitinib 42-51 AXL receptor tyrosine kinase Homo sapiens 13-16 28327411-3 2017 Gas6-induced Axl signaling in presence of Sunitinib is also diversified displaying onset of Axl-dependent EGFR and METR activation and activation of classical MAPK pathways. Sunitinib 42-51 epidermal growth factor receptor Homo sapiens 106-110 28327411-4 2017 Gas6+Sunitinib-adapted CCRCC cells present increased viability and decreased apoptosis and enhanced production of the multi-tumorigenic Osteopontin (OPN) and of one of its activator matrix metalloproteinase-7. Sunitinib 5-14 growth arrest specific 6 Homo sapiens 0-4 28327411-4 2017 Gas6+Sunitinib-adapted CCRCC cells present increased viability and decreased apoptosis and enhanced production of the multi-tumorigenic Osteopontin (OPN) and of one of its activator matrix metalloproteinase-7. Sunitinib 5-14 secreted phosphoprotein 1 Homo sapiens 136-147 28327411-4 2017 Gas6+Sunitinib-adapted CCRCC cells present increased viability and decreased apoptosis and enhanced production of the multi-tumorigenic Osteopontin (OPN) and of one of its activator matrix metalloproteinase-7. Sunitinib 5-14 secreted phosphoprotein 1 Homo sapiens 149-152 28327411-4 2017 Gas6+Sunitinib-adapted CCRCC cells present increased viability and decreased apoptosis and enhanced production of the multi-tumorigenic Osteopontin (OPN) and of one of its activator matrix metalloproteinase-7. Sunitinib 5-14 matrix metallopeptidase 7 Homo sapiens 182-208 28327411-6 2017 In addition, Gas6+Sunitinib-adapted CCRCC cells displayed enhanced migration and sphere formation, both mechanisms being Axl and OPN dependent. Sunitinib 18-27 growth arrest specific 6 Homo sapiens 13-17 28327411-6 2017 In addition, Gas6+Sunitinib-adapted CCRCC cells displayed enhanced migration and sphere formation, both mechanisms being Axl and OPN dependent. Sunitinib 18-27 AXL receptor tyrosine kinase Homo sapiens 121-124 28327411-6 2017 In addition, Gas6+Sunitinib-adapted CCRCC cells displayed enhanced migration and sphere formation, both mechanisms being Axl and OPN dependent. Sunitinib 18-27 secreted phosphoprotein 1 Homo sapiens 129-132 28327411-7 2017 Altogether, this suggests that Sunitinib while targeting endothelial cells and tumor angiogenesis, simultaneously provides protumorigenic effects due to a constitutively, intensified and divergent Gas6-Axl system. Sunitinib 31-40 growth arrest specific 6 Homo sapiens 197-201 28327411-7 2017 Altogether, this suggests that Sunitinib while targeting endothelial cells and tumor angiogenesis, simultaneously provides protumorigenic effects due to a constitutively, intensified and divergent Gas6-Axl system. Sunitinib 31-40 AXL receptor tyrosine kinase Homo sapiens 202-205 28327411-8 2017 IMPLICATIONS: Gas6-mediated Axl signaling, which is enhanced and diversified in the presence of Sunitinib possibly contributes to acquired chemoresistance, recurrence of aggressive disease and metastasis of CCRCC tumors. Sunitinib 96-105 growth arrest specific 6 Homo sapiens 14-18 28327411-8 2017 IMPLICATIONS: Gas6-mediated Axl signaling, which is enhanced and diversified in the presence of Sunitinib possibly contributes to acquired chemoresistance, recurrence of aggressive disease and metastasis of CCRCC tumors. Sunitinib 96-105 AXL receptor tyrosine kinase Homo sapiens 28-31 28327411-9 2017 Therefore, combinatorial Axl-targeted therapy might be beneficial for CCRCC patients intended for Sunitinib treatment. Sunitinib 98-107 AXL receptor tyrosine kinase Homo sapiens 25-28 28736633-3 2017 Through a number of clinical trials, the mTOR inhibitor everolimus and the receptor tyrosine kinase (RTK) inhibitor sunitinib were recently approved for NETs. Sunitinib 116-125 ret proto-oncogene Homo sapiens 75-99 28260162-0 2017 Tumoral cubilin is a predictive marker for treatment of renal cancer patients with sunitinib and sorafenib. Sunitinib 83-92 cubilin Homo sapiens 8-15 28260162-12 2017 CONCLUSIONS: We show for the first time that tumoral expression of cubilin is a positive predictive marker for treatment of metastatic renal cell cancer patients with sunitinib and sorafenib. Sunitinib 167-176 cubilin Homo sapiens 67-74 28736633-3 2017 Through a number of clinical trials, the mTOR inhibitor everolimus and the receptor tyrosine kinase (RTK) inhibitor sunitinib were recently approved for NETs. Sunitinib 116-125 ret proto-oncogene Homo sapiens 101-104 28599497-7 2017 In A549-xenografted nude mice treated with sunitinib or cetuximab, a decrease in the plasma AMF concentration was accompanied by a reduction in tumor weight, suggesting an association between the plasma AMF concentration and anticancer activity. Sunitinib 43-52 glucose-6-phosphate isomerase Homo sapiens 92-95 28452850-0 2017 Sunitinib Induces NK-kappaB-dependent NKG2D Ligand Expression in Nasopharyngeal Carcinoma and Hepatoma Cells. Sunitinib 0-9 killer cell lectin like receptor C1 Homo sapiens 38-42 28452850-4 2017 In this study, we confirmed sunitinib induced downregulation of its targets, such as vascular endothelial growth factor, platelet-derived growth factor, and c-kit in multiple-drug-resistant nasopharyngeal carcinoma cell line CNE2/DDP and hepatoma cell line HepG2. Sunitinib 28-37 vascular endothelial growth factor A Homo sapiens 85-119 28452850-4 2017 In this study, we confirmed sunitinib induced downregulation of its targets, such as vascular endothelial growth factor, platelet-derived growth factor, and c-kit in multiple-drug-resistant nasopharyngeal carcinoma cell line CNE2/DDP and hepatoma cell line HepG2. Sunitinib 28-37 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 157-162 28452850-8 2017 Silencing of NF-kappabeta1, NF-kappabeta2, or RelB (NF-kappabeta pathway) inhibited sunitinib-induced upregulation of NKG2DLs. Sunitinib 84-93 RELB proto-oncogene, NF-kB subunit Homo sapiens 46-50 28452850-8 2017 Silencing of NF-kappabeta1, NF-kappabeta2, or RelB (NF-kappabeta pathway) inhibited sunitinib-induced upregulation of NKG2DLs. Sunitinib 84-93 nuclear factor kappa B subunit 1 Homo sapiens 13-25 28452850-9 2017 Taken together, we concluded that sunitinib upregulated NKG2DLs through NF-kappabeta signaling noncanonical pathway which might mediate higher cytotoxic sensitivity of CNE2/DDP and HepG2 cells to NK cells. Sunitinib 34-43 nuclear factor kappa B subunit 1 Homo sapiens 72-84 28602541-0 2017 Connexin 43 enhances Bax activation via JNK activation in sunitinib-induced apoptosis in mesothelioma cells. Sunitinib 58-67 gap junction protein alpha 1 Homo sapiens 0-11 28602541-0 2017 Connexin 43 enhances Bax activation via JNK activation in sunitinib-induced apoptosis in mesothelioma cells. Sunitinib 58-67 BCL2 associated X, apoptosis regulator Homo sapiens 21-24 28602541-0 2017 Connexin 43 enhances Bax activation via JNK activation in sunitinib-induced apoptosis in mesothelioma cells. Sunitinib 58-67 mitogen-activated protein kinase 8 Homo sapiens 40-43 28602541-3 2017 In our previous work, Cx43 was found to interact directly with Bax and in the presence of sunitinib, lead to the Bax-mediated apoptosis in mesothelioma cells. Sunitinib 90-99 gap junction protein alpha 1 Homo sapiens 22-26 28602541-3 2017 In our previous work, Cx43 was found to interact directly with Bax and in the presence of sunitinib, lead to the Bax-mediated apoptosis in mesothelioma cells. Sunitinib 90-99 BCL2 associated X, apoptosis regulator Homo sapiens 113-116 28602541-5 2017 Treatment with sunitinib increased the expression of the active conformation of the Bax protein, which was predominantly localized at the mitochondria, only in Cx43-transfected cells. Sunitinib 15-24 BCL2 associated X, apoptosis regulator Homo sapiens 84-87 28602541-5 2017 Treatment with sunitinib increased the expression of the active conformation of the Bax protein, which was predominantly localized at the mitochondria, only in Cx43-transfected cells. Sunitinib 15-24 gap junction protein alpha 1 Homo sapiens 160-164 28602541-8 2017 Treatment with sunitinib increased the expression of phosphorylated (active) form of JNK only in the Cx43-transfected cells. Sunitinib 15-24 mitogen-activated protein kinase 8 Homo sapiens 85-88 28602541-8 2017 Treatment with sunitinib increased the expression of phosphorylated (active) form of JNK only in the Cx43-transfected cells. Sunitinib 15-24 gap junction protein alpha 1 Homo sapiens 101-105 28599497-7 2017 In A549-xenografted nude mice treated with sunitinib or cetuximab, a decrease in the plasma AMF concentration was accompanied by a reduction in tumor weight, suggesting an association between the plasma AMF concentration and anticancer activity. Sunitinib 43-52 glucose-6-phosphate isomerase Homo sapiens 203-206 28330784-8 2017 Small molecule VEGFR1/2/3 inhibitors including axitinib, cabozantinib, lenvatinib, sorafenib, sunitinib, and pazopanib are approved by the FDA for the treatment of renal cell carcinomas. Sunitinib 94-103 fms related receptor tyrosine kinase 1 Homo sapiens 15-25 28561780-4 2017 Of these, 11 and 12 had the highest potency and, compared to sunitinib, showed: (1) significant increase in anti-proliferation of various cancer cells with a favorable selective index (SI); (2) higher inhibitory potency against both VEGFR-2 and PDGFRbeta. Sunitinib 61-70 kinase insert domain receptor Homo sapiens 233-240 28430711-0 2017 Evaluation of KDR rs34231037 as a predictor of sunitinib efficacy in patients with metastatic renal cell carcinoma. Sunitinib 47-56 kinase insert domain receptor Homo sapiens 14-17 28430711-5 2017 KDR-rs34231037 association with sunitinib response, clinical benefit, and progression-free survival was analyzed using logistic and Cox regression analyses. Sunitinib 32-41 kinase insert domain receptor Homo sapiens 0-3 28478525-3 2017 Three tyrosine kinase inhibitors (TKIs) with KIT inhibitory activity - imatinib, sunitinib, and regorafenib - are approved to treat advanced GIST and have successfully exploited this addiction to KIT oncogenic signaling, demonstrating remarkable activity in a disease that historically had no successful systemic therapy options. Sunitinib 81-90 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 196-199 28561780-5 2017 The molecular modeling results showed that, in terms of VEGFR-2 binding, the synthesized products had a similar binding mode to sunitinib but with tighter interaction. Sunitinib 128-137 kinase insert domain receptor Homo sapiens 56-63 28529744-0 2017 Prognostic effect of serum C-reactive protein kinetics on advanced renal cell carcinoma treated with sunitinib. Sunitinib 101-110 C-reactive protein Homo sapiens 27-45 28205224-5 2017 Silencing of MUC13 expression inhibited migration and invasion, and sensitized renal cancer cells to killing by the multi-kinase inhibitors used clinically, sorafenib and sunitinib, and reversed acquired resistance to these drugs. Sunitinib 171-180 mucin 13, cell surface associated Homo sapiens 13-18 28423742-15 2017 HEYL, hsa-miR-27b, hsa-miR-23b and hsa-miR-628-5p could be potentially used as biomarkers of sunitinib response. Sunitinib 93-102 hes related family bHLH transcription factor with YRPW motif like Homo sapiens 0-4 28423742-15 2017 HEYL, hsa-miR-27b, hsa-miR-23b and hsa-miR-628-5p could be potentially used as biomarkers of sunitinib response. Sunitinib 93-102 microRNA 27b Homo sapiens 10-17 28423742-15 2017 HEYL, hsa-miR-27b, hsa-miR-23b and hsa-miR-628-5p could be potentially used as biomarkers of sunitinib response. Sunitinib 93-102 microRNA 628 Homo sapiens 35-46 28529744-2 2017 The aim of the present study was to investigate the prognostic effect of pretreatment serum CRP level and CRP kinetics on patients with advanced RCC treated with sunitinib. Sunitinib 162-171 C-reactive protein Homo sapiens 106-109 28529744-9 2017 Therefore, CRP kinetics and normal pretreatment CRP level are prognostic indicators in patients with advanced RCC treated with sunitinib. Sunitinib 127-136 C-reactive protein Homo sapiens 11-14 28529744-2 2017 The aim of the present study was to investigate the prognostic effect of pretreatment serum CRP level and CRP kinetics on patients with advanced RCC treated with sunitinib. Sunitinib 162-171 C-reactive protein Homo sapiens 92-95 28529744-9 2017 Therefore, CRP kinetics and normal pretreatment CRP level are prognostic indicators in patients with advanced RCC treated with sunitinib. Sunitinib 127-136 C-reactive protein Homo sapiens 48-51 28418873-6 2017 Besides, combined treatment with sunitinib, PF-04691502 and GANT61 significantly prolonged the survival of mice transplanted with FLT3-mutated MV4-11 cells compared to the single agent treatments. Sunitinib 33-42 FMS-like tyrosine kinase 3 Mus musculus 130-134 28670632-1 2017 PURPOSE: To describe the clinical course of advanced juxtapapillary retinal capillary hemangioblastomas (RCH) associated with von Hippel-Lindau (VHL) disease treated with systemic sunitinib malate, an agent that inhibits both anti-vascular endothelial growth factor and anti-platelet-derived growth factor signaling. Sunitinib 180-196 vascular endothelial growth factor A Homo sapiens 231-265 28105557-3 2017 Although no treatments are currently available to manage HAND, we have previously shown that sunitinib, an anticancer drug that blocks receptor tyrosine-kinase and cyclin kinase pathways, might be of interest. Sunitinib 93-102 ret proto-oncogene Homo sapiens 135-159 28240606-6 2017 In cultured cells, treatment with sunitinib and erlotinib, approved anticancer drugs that inhibit AAK1 or GAK activity, or with more selective compounds inhibited intracellular trafficking of HCV and multiple unrelated RNA viruses with a high barrier to resistance. Sunitinib 34-43 AP2 associated kinase 1 Homo sapiens 98-102 28240606-6 2017 In cultured cells, treatment with sunitinib and erlotinib, approved anticancer drugs that inhibit AAK1 or GAK activity, or with more selective compounds inhibited intracellular trafficking of HCV and multiple unrelated RNA viruses with a high barrier to resistance. Sunitinib 34-43 cyclin G associated kinase Homo sapiens 106-109 28240606-8 2017 We validated sunitinib- and erlotinib-mediated inhibition of AAK1 and GAK activity as an important mechanism of antiviral action. Sunitinib 13-22 AP2 associated kinase 1 Homo sapiens 61-65 28240606-8 2017 We validated sunitinib- and erlotinib-mediated inhibition of AAK1 and GAK activity as an important mechanism of antiviral action. Sunitinib 13-22 cyclin G associated kinase Homo sapiens 70-73 28413468-6 2017 In addition, sunitinib activated ERK1/2 and inhibited mTOR/p70S6K signaling. Sunitinib 13-22 mitogen-activated protein kinase 3 Homo sapiens 33-39 28413468-6 2017 In addition, sunitinib activated ERK1/2 and inhibited mTOR/p70S6K signaling. Sunitinib 13-22 mechanistic target of rapamycin kinase Homo sapiens 54-58 28413468-6 2017 In addition, sunitinib activated ERK1/2 and inhibited mTOR/p70S6K signaling. Sunitinib 13-22 ribosomal protein S6 kinase B1 Homo sapiens 59-65 28413468-7 2017 Sunitinib-induced autophagy was notably reversed by ERK1/2 kinase inhibitor, U0126. Sunitinib 0-9 mitogen-activated protein kinase 3 Homo sapiens 52-58 28105557-8 2017 Moreover, in these animals, sunitinib reduced the hyperactivation of CDK5, tau hyperphosphorylation, and p35 cleavage to p25. Sunitinib 28-37 cyclin dependent kinase 5 regulatory subunit 1 Homo sapiens 121-124 28105557-10 2017 Alterations in autophagy in the Tat tg mice were associated with reduced levels of a CDK5 substrate, EndoB1, and levels of total EndoB1 were normalized by sunitinib treatment. Sunitinib 155-164 cyclin-dependent kinase 5 Mus musculus 85-89 28105557-10 2017 Alterations in autophagy in the Tat tg mice were associated with reduced levels of a CDK5 substrate, EndoB1, and levels of total EndoB1 were normalized by sunitinib treatment. Sunitinib 155-164 SH3-domain GRB2-like B1 (endophilin) Mus musculus 101-107 28105557-10 2017 Alterations in autophagy in the Tat tg mice were associated with reduced levels of a CDK5 substrate, EndoB1, and levels of total EndoB1 were normalized by sunitinib treatment. Sunitinib 155-164 SH3-domain GRB2-like B1 (endophilin) Mus musculus 129-135 28384190-12 2017 HPLC analysis of the cell culture supernatant demonstrated saturation of the cell medium within approximately 4 hours for each amount added, with sunitinib achieving a final concentration of 17.61 muM (SE +-1.01). Sunitinib 146-155 latexin Homo sapiens 197-200 28278077-12 2017 Moreover, the VM-associated proteins VE-cadherin and Twist1 upregulated in the sunitinib-treated MDA-MB-231 and Hs578T tumors. Sunitinib 79-88 cadherin 5 Homo sapiens 37-48 28278077-12 2017 Moreover, the VM-associated proteins VE-cadherin and Twist1 upregulated in the sunitinib-treated MDA-MB-231 and Hs578T tumors. Sunitinib 79-88 twist family bHLH transcription factor 1 Homo sapiens 53-59 28105557-6 2017 In neuronal cultures challenged with low levels of Tat, sunitinib increased markers of autophagy such as LC3-II and reduced p62 accumulation in a dose-dependent manner. Sunitinib 56-65 nucleoporin 62 Homo sapiens 124-127 28105557-7 2017 In vivo, sunitinib treatment restored LC3-II, p62, and endophilin B1 (EndoB1) levels in doxycycline-induced Tat transgenic mice. Sunitinib 9-18 nucleoporin 62 Mus musculus 46-49 28105557-7 2017 In vivo, sunitinib treatment restored LC3-II, p62, and endophilin B1 (EndoB1) levels in doxycycline-induced Tat transgenic mice. Sunitinib 9-18 SH3-domain GRB2-like B1 (endophilin) Mus musculus 55-68 28105557-7 2017 In vivo, sunitinib treatment restored LC3-II, p62, and endophilin B1 (EndoB1) levels in doxycycline-induced Tat transgenic mice. Sunitinib 9-18 SH3-domain GRB2-like B1 (endophilin) Mus musculus 70-76 28105557-8 2017 Moreover, in these animals, sunitinib reduced the hyperactivation of CDK5, tau hyperphosphorylation, and p35 cleavage to p25. Sunitinib 28-37 cyclin dependent kinase 5 Homo sapiens 69-73 28105557-8 2017 Moreover, in these animals, sunitinib reduced the hyperactivation of CDK5, tau hyperphosphorylation, and p35 cleavage to p25. Sunitinib 28-37 cyclin dependent kinase 5 regulatory subunit 1 Homo sapiens 105-108 28274318-0 2017 [Knockdown of ATG5 enhances the sensitivity of human renal carcinoma cells to sunitinib]. Sunitinib 78-87 autophagy related 5 Homo sapiens 14-18 28423517-6 2017 The expression levels of sunitinib targeted-kinases were measured by transcriptome sequencing for KDR, PDGFRA/B, KIT, RET, FLT1, and FLT4. Sunitinib 25-34 kinase insert domain receptor Homo sapiens 98-101 28184014-8 2017 These results were confirmed by siRNA targeting the autophagy-related gene Atg5 In CCOC tumor xenografts, Lys05 potentiated the antitumor activity of sunitinib compared with either treatment alone. Sunitinib 150-159 autophagy related 5 Homo sapiens 75-79 28231773-2 2017 METHODS: Eighteen patients with pathologically proven advanced GEP-NENs treated with sunitinib were enrolled in the study. Sunitinib 85-94 granulin precursor Homo sapiens 63-66 28231773-13 2017 CONCLUSION: The Choi criteria appear to be more sensitive and more precise than RECIST 1.1 in assessing the early response of advanced GEP-NENs treated with sunitinib. Sunitinib 157-166 granulin precursor Homo sapiens 135-138 28401187-3 2017 Inhibition of MCL-1 or mTORC1 sensitized cancer cells to sunitinib. Sunitinib 57-66 MCL1 apoptosis regulator, BCL2 family member Homo sapiens 14-19 28167241-7 2017 Sunitinib treatment reduced vessel densities and induced hypoxia in all melanoma lines, and the magnitude of the effect was associated with the gene expression and protein secretion rate of VEGF-A. Sunitinib 0-9 vascular endothelial growth factor A Homo sapiens 190-196 28276433-5 2017 However, over the past decade, marked advances in the treatment of metastatic RCC have been made, with targeted agents including sorafenib, sunitinib, bevacizumab, pazopanib and axitinib, which inhibit vascular endothelial growth factor (VEGF) and its receptor (VEGFR), and everolimus and temsirolimus, which inhibit mechanistic target of rapamycin complex 1 (mTORC1), being approved. Sunitinib 140-149 vascular endothelial growth factor A Homo sapiens 202-236 28276433-5 2017 However, over the past decade, marked advances in the treatment of metastatic RCC have been made, with targeted agents including sorafenib, sunitinib, bevacizumab, pazopanib and axitinib, which inhibit vascular endothelial growth factor (VEGF) and its receptor (VEGFR), and everolimus and temsirolimus, which inhibit mechanistic target of rapamycin complex 1 (mTORC1), being approved. Sunitinib 140-149 vascular endothelial growth factor A Homo sapiens 238-242 28276433-5 2017 However, over the past decade, marked advances in the treatment of metastatic RCC have been made, with targeted agents including sorafenib, sunitinib, bevacizumab, pazopanib and axitinib, which inhibit vascular endothelial growth factor (VEGF) and its receptor (VEGFR), and everolimus and temsirolimus, which inhibit mechanistic target of rapamycin complex 1 (mTORC1), being approved. Sunitinib 140-149 kinase insert domain receptor Homo sapiens 262-267 28276433-5 2017 However, over the past decade, marked advances in the treatment of metastatic RCC have been made, with targeted agents including sorafenib, sunitinib, bevacizumab, pazopanib and axitinib, which inhibit vascular endothelial growth factor (VEGF) and its receptor (VEGFR), and everolimus and temsirolimus, which inhibit mechanistic target of rapamycin complex 1 (mTORC1), being approved. Sunitinib 140-149 CREB regulated transcription coactivator 1 Mus musculus 360-366 28087600-0 2017 Sunitinib Stimulates Expression of VEGFC by Tumor Cells and Promotes Lymphangiogenesis in Clear Cell Renal Cell Carcinomas. Sunitinib 0-9 vascular endothelial growth factor C Homo sapiens 35-40 28087600-4 2017 In this study, we found that sunitinib can stimulate vegfc gene transcription and increase VEGFC mRNA half-life. Sunitinib 29-38 vascular endothelial growth factor C Homo sapiens 53-58 28087600-4 2017 In this study, we found that sunitinib can stimulate vegfc gene transcription and increase VEGFC mRNA half-life. Sunitinib 29-38 vascular endothelial growth factor C Homo sapiens 91-96 28087600-5 2017 In addition, sunitinib activated p38 MAPK, which resulted in the upregulation/activity of HuR and inactivation of tristetraprolin, two AU-rich element-binding proteins. Sunitinib 13-22 ELAV like RNA binding protein 1 Homo sapiens 90-93 28087600-5 2017 In addition, sunitinib activated p38 MAPK, which resulted in the upregulation/activity of HuR and inactivation of tristetraprolin, two AU-rich element-binding proteins. Sunitinib 13-22 ZFP36 ring finger protein Homo sapiens 114-129 28087600-6 2017 Sunitinib stimulated a VEGFC-dependent development of lymphatic vessels in experimental tumors. Sunitinib 0-9 vascular endothelial growth factor C Homo sapiens 23-28 27751729-8 2017 With first-line sunitinib, KDM5C mutations were associated with longer PFS1L (median [95% CI] of 20.6 [12.4, 27.3] vs 8.3 [7.8, 11.0] mo). Sunitinib 16-25 lysine demethylase 5C Homo sapiens 27-32 27395374-1 2017 PURPOSE: To identify prognostic molecular profiles in patients with mRCC treated with sunitinib, we performed immunohistochemical analysis for VEGF and PI3K/Akt/mTOR pathway components. Sunitinib 86-95 vascular endothelial growth factor A Homo sapiens 143-147 27395374-7 2017 CONCLUSION: Immunohistochemistry for VEGF and p-mTOR proteins may discriminate patients refractory to first-line sunitinib with poor prognosis. Sunitinib 113-122 vascular endothelial growth factor A Homo sapiens 37-41 27395374-7 2017 CONCLUSION: Immunohistochemistry for VEGF and p-mTOR proteins may discriminate patients refractory to first-line sunitinib with poor prognosis. Sunitinib 113-122 mechanistic target of rapamycin kinase Homo sapiens 48-52 27477693-7 2017 Taken together, our study suggests a model in which overexpression of wild-type PDGFRA associated with NF1 deficiency leads to aberrant activation of downstream RAS signaling and thus contributes importantly to MPNST development-a prediction supported by the ability of the kinase inhibitor sunitinib alone and in combination with the MEK inhibitor trametinib to retard MPNST progression in transgenic fish overexpressing the wild-type receptor. Sunitinib 291-300 platelet-derived growth factor receptor, alpha polypeptide Danio rerio 80-86 28401187-3 2017 Inhibition of MCL-1 or mTORC1 sensitized cancer cells to sunitinib. Sunitinib 57-66 CREB regulated transcription coactivator 1 Mus musculus 23-29 28401187-4 2017 Analysis of tissues from patients correlated MCL-1/mTORC1 induction with resistance to sunitinib. Sunitinib 87-96 MCL1 apoptosis regulator, BCL2 family member Homo sapiens 45-50 28401187-4 2017 Analysis of tissues from patients correlated MCL-1/mTORC1 induction with resistance to sunitinib. Sunitinib 87-96 CREB regulated transcription coactivator 1 Mus musculus 51-57 28274318-1 2017 Objective To investigate the expression levels of autophagy-related gene 5 (ATG5) and microtubule-associated protein 1 light chain 3 (LC3) and their effects on sunitinib resistance in human renal carcinoma cells. Sunitinib 160-169 autophagy related 5 Homo sapiens 50-74 28274318-1 2017 Objective To investigate the expression levels of autophagy-related gene 5 (ATG5) and microtubule-associated protein 1 light chain 3 (LC3) and their effects on sunitinib resistance in human renal carcinoma cells. Sunitinib 160-169 autophagy related 5 Homo sapiens 76-80 28274318-1 2017 Objective To investigate the expression levels of autophagy-related gene 5 (ATG5) and microtubule-associated protein 1 light chain 3 (LC3) and their effects on sunitinib resistance in human renal carcinoma cells. Sunitinib 160-169 microtubule associated protein 1 light chain 3 alpha Homo sapiens 134-137 28274318-11 2017 Compared with control group, the survival rate in ATG5 low expression group were significantly reduced in a dose-dependent manner after sunitinib treatment. Sunitinib 136-145 autophagy related 5 Homo sapiens 50-54 28274318-12 2017 Conclusion Autophagy is active in renal clear cell carcinoma, and the drug sensitivity to sunitinib in renal cancer cells can be enhanced by the downregulation of ATG5. Sunitinib 90-99 autophagy related 5 Homo sapiens 163-167 27896475-9 2017 We also highlighted the association with sunitinib-related toxicity; in particular, VEGFA polymorphism rs3025039 (CT+TT vs. CC, OR 15.3, 95% CI 2.2-102.1; P = 0.005) is associated with severe toxicity, with the presence of the variant T allele associated with a grade >=3 AE. Sunitinib 41-50 vascular endothelial growth factor A Homo sapiens 84-89 28143610-10 2017 In this case, the patient was homozygous for the ABCG2 421C allele, but was capable of potentially harboring polymorphisms in other genes, such as ABCB1, an efflux pump of sunitinib. Sunitinib 172-181 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 49-54 27030134-1 2017 AIM: Although targeting angiogenesis with vascular endothelial growth factor receptor (VEGFR) inhibitors has demonstrated efficacy in the treatment of renal cell carcinoma, primary, intrinsic resistance to the VEGFR inhibitor sunitinib is not fully understood in a subset of metastatic RCC (mRCC) patients. Sunitinib 226-235 kinase insert domain receptor Homo sapiens 42-85 27030134-1 2017 AIM: Although targeting angiogenesis with vascular endothelial growth factor receptor (VEGFR) inhibitors has demonstrated efficacy in the treatment of renal cell carcinoma, primary, intrinsic resistance to the VEGFR inhibitor sunitinib is not fully understood in a subset of metastatic RCC (mRCC) patients. Sunitinib 226-235 kinase insert domain receptor Homo sapiens 87-92 27030134-1 2017 AIM: Although targeting angiogenesis with vascular endothelial growth factor receptor (VEGFR) inhibitors has demonstrated efficacy in the treatment of renal cell carcinoma, primary, intrinsic resistance to the VEGFR inhibitor sunitinib is not fully understood in a subset of metastatic RCC (mRCC) patients. Sunitinib 226-235 kinase insert domain receptor Homo sapiens 210-215 28143610-10 2017 In this case, the patient was homozygous for the ABCG2 421C allele, but was capable of potentially harboring polymorphisms in other genes, such as ABCB1, an efflux pump of sunitinib. Sunitinib 172-181 ATP binding cassette subfamily B member 1 Homo sapiens 147-152 28117391-0 2017 Functional PTGS2 polymorphism-based models as novel predictive markers in metastatic renal cell carcinoma patients receiving first-line sunitinib. Sunitinib 136-145 prostaglandin-endoperoxide synthase 2 Homo sapiens 11-16 28217462-5 2017 In conjunction with the restricted expression of HIF-2alpha in normal adult physiology, these studies suggest that such therapeutic approach might be favorable for patients with lower toxicity than current anti-angiogenic drugs like sunitinib. Sunitinib 233-242 endothelial PAS domain protein 1 Homo sapiens 49-59 28114889-3 2017 METHODS: In this study, in a clinical aspect, we retrospectively investigated the prognostic and predictive impact of tumoral CCR7 expression in 110 mRCC patients treated with sunitinib and sorafenib, and its correlation with pre- or post-administration lymphatic involvement. Sunitinib 176-185 C-C motif chemokine receptor 7 Homo sapiens 126-130 28178124-1 2017 RATIONALE: The case reported the rapid remission of disease recurrence achieved adding foscarnet, a DNA polymerase inhibitor that interacts with fibroblast growth factor 2, to low molecular weight heparin and sunitinib for the first time in a patient with an anaplastic thyroid cancer (ATC). Sunitinib 209-218 fibroblast growth factor 2 Homo sapiens 145-171 27958294-0 2017 Targeted therapy: Sunitinib modulates MCL-1 and mTOR signalling. Sunitinib 18-27 MCL1 apoptosis regulator, BCL2 family member Homo sapiens 38-43 27958294-0 2017 Targeted therapy: Sunitinib modulates MCL-1 and mTOR signalling. Sunitinib 18-27 mechanistic target of rapamycin kinase Homo sapiens 48-52 27893461-0 2017 Dual modulation of MCL-1 and mTOR determines the response to sunitinib. Sunitinib 61-70 MCL1 apoptosis regulator, BCL2 family member Homo sapiens 19-24 28117425-2 2017 The endothelial cell-specific mitogen vascular endothelial growth factor (VEGF), as well as its receptors, VEGFR1, VEGFR2, are thought to be the major mediators of pathological angiogenesis, and sunitinib exhibits anti-angiogenesis property through VEGF blockage and has been widely used to treat various cancers. Sunitinib 195-204 vascular endothelial growth factor A Rattus norvegicus 38-72 28117425-2 2017 The endothelial cell-specific mitogen vascular endothelial growth factor (VEGF), as well as its receptors, VEGFR1, VEGFR2, are thought to be the major mediators of pathological angiogenesis, and sunitinib exhibits anti-angiogenesis property through VEGF blockage and has been widely used to treat various cancers. Sunitinib 195-204 vascular endothelial growth factor A Rattus norvegicus 74-78 28117425-2 2017 The endothelial cell-specific mitogen vascular endothelial growth factor (VEGF), as well as its receptors, VEGFR1, VEGFR2, are thought to be the major mediators of pathological angiogenesis, and sunitinib exhibits anti-angiogenesis property through VEGF blockage and has been widely used to treat various cancers. Sunitinib 195-204 Fms related receptor tyrosine kinase 1 Rattus norvegicus 107-113 28117425-2 2017 The endothelial cell-specific mitogen vascular endothelial growth factor (VEGF), as well as its receptors, VEGFR1, VEGFR2, are thought to be the major mediators of pathological angiogenesis, and sunitinib exhibits anti-angiogenesis property through VEGF blockage and has been widely used to treat various cancers. Sunitinib 195-204 kinase insert domain receptor Rattus norvegicus 115-121 28117425-2 2017 The endothelial cell-specific mitogen vascular endothelial growth factor (VEGF), as well as its receptors, VEGFR1, VEGFR2, are thought to be the major mediators of pathological angiogenesis, and sunitinib exhibits anti-angiogenesis property through VEGF blockage and has been widely used to treat various cancers. Sunitinib 195-204 vascular endothelial growth factor A Rattus norvegicus 107-111 28117425-5 2017 Sunitinib treatment was associated with less angiogenesis in small-airway remodelling with a slightly disordered lung architecture, and lower expression level of VEGF, VEGFR1, VEGFR2. Sunitinib 0-9 vascular endothelial growth factor A Rattus norvegicus 162-166 28117425-5 2017 Sunitinib treatment was associated with less angiogenesis in small-airway remodelling with a slightly disordered lung architecture, and lower expression level of VEGF, VEGFR1, VEGFR2. Sunitinib 0-9 Fms related receptor tyrosine kinase 1 Rattus norvegicus 168-174 28117425-5 2017 Sunitinib treatment was associated with less angiogenesis in small-airway remodelling with a slightly disordered lung architecture, and lower expression level of VEGF, VEGFR1, VEGFR2. Sunitinib 0-9 kinase insert domain receptor Rattus norvegicus 176-182 28117425-6 2017 Overall, our results indicate that VEGF is a vital important factor that contributes to the small-airway remodelling in a rat model of COPD through promoting angiogenesis, which mainly depend on the specific binding between VEGF and VEGFR1 and can be effectively attenuated by sunitinib. Sunitinib 277-286 vascular endothelial growth factor A Rattus norvegicus 35-39 28117425-6 2017 Overall, our results indicate that VEGF is a vital important factor that contributes to the small-airway remodelling in a rat model of COPD through promoting angiogenesis, which mainly depend on the specific binding between VEGF and VEGFR1 and can be effectively attenuated by sunitinib. Sunitinib 277-286 vascular endothelial growth factor A Rattus norvegicus 224-228 28117425-6 2017 Overall, our results indicate that VEGF is a vital important factor that contributes to the small-airway remodelling in a rat model of COPD through promoting angiogenesis, which mainly depend on the specific binding between VEGF and VEGFR1 and can be effectively attenuated by sunitinib. Sunitinib 277-286 Fms related receptor tyrosine kinase 1 Rattus norvegicus 233-239 27997164-0 2017 Synthesis of Novel c(AmpRGD)-Sunitinib Dual Conjugates as Molecular Tools Targeting the alphavbeta3 Integrin/VEGFR2 Couple and Impairing Tumor-Associated Angiogenesis. Sunitinib 29-38 kinase insert domain protein receptor Mus musculus 109-115 28057017-6 2017 RESULTS: The in vivo experiments demonstrated that bevacizumab and sunitinib increase the in vivo expression of CXCR4, SDF-1alpha, and TGFbeta1. Sunitinib 67-76 C-X-C motif chemokine receptor 4 Homo sapiens 112-117 28057017-6 2017 RESULTS: The in vivo experiments demonstrated that bevacizumab and sunitinib increase the in vivo expression of CXCR4, SDF-1alpha, and TGFbeta1. Sunitinib 67-76 transforming growth factor beta 1 Homo sapiens 135-143 28057017-7 2017 In addition, we demonstrate that the co-administration of the novel brain-penetrating CXCR4 antagonist, PRX177561, with bevacizumab or sunitinib inhibited tumor growth and reduced the inflammation. Sunitinib 135-144 C-X-C motif chemokine receptor 4 Homo sapiens 86-91 28057017-0 2017 The brain-penetrating CXCR4 antagonist, PRX177561, increases the antitumor effects of bevacizumab and sunitinib in preclinical models of human glioblastoma. Sunitinib 102-111 C-X-C motif chemokine receptor 4 Homo sapiens 22-27 27893461-0 2017 Dual modulation of MCL-1 and mTOR determines the response to sunitinib. Sunitinib 61-70 mechanistic target of rapamycin kinase Homo sapiens 29-33 27901483-0 2017 Meta-analysis on the association of VEGFR1 genetic variants with sunitinib outcome in metastatic renal cell carcinoma patients. Sunitinib 65-74 fms related receptor tyrosine kinase 1 Homo sapiens 36-42 27893461-3 2017 Here, we have shown that cancer cells respond to clinically relevant doses of sunitinib by enhancing the stability of the antiapoptotic protein MCL-1 and inducing mTORC1 signaling, thus evoking little cytotoxicity. Sunitinib 78-87 MCL1 apoptosis regulator, BCL2 family member Homo sapiens 144-149 27901483-1 2017 VEGFR1 rs9582036 and rs9554320 were previously reported the association with sunitinib progression-free survival (PFS) and overall survival (OS) in patients with metastatic renal cell carcinoma (mRCC). Sunitinib 77-86 fms related receptor tyrosine kinase 1 Homo sapiens 0-6 27901483-8 2017 Our findings suggest that, although VEGFR1 rs9582036 and rs9554320 are involved in sunitinib therapy outcome, its clinical use as biomarkers for prediction of sunitinib outcome in mRCC patients is limited, due to inconsistent findings when analyzing all existing studies together. Sunitinib 83-92 fms related receptor tyrosine kinase 1 Homo sapiens 36-42 27893461-3 2017 Here, we have shown that cancer cells respond to clinically relevant doses of sunitinib by enhancing the stability of the antiapoptotic protein MCL-1 and inducing mTORC1 signaling, thus evoking little cytotoxicity. Sunitinib 78-87 CREB regulated transcription coactivator 1 Mus musculus 163-169 27926489-9 2017 The expression of GM-CSF was substantially inhibited by celecoxib and sunitinib. Sunitinib 70-79 colony stimulating factor 2 Homo sapiens 18-24 27893461-4 2017 Inhibition of MCL-1 or mTORC1 signaling sensitized cells to clinically relevant doses of sunitinib in vitro and was synergistic with sunitinib in impairing tumor growth in vivo, indicating that these responses are triggered as prosurvival mechanisms that enable cells to tolerate the cytotoxic effects of sunitinib. Sunitinib 89-98 MCL1 apoptosis regulator, BCL2 family member Homo sapiens 14-19 27893461-4 2017 Inhibition of MCL-1 or mTORC1 signaling sensitized cells to clinically relevant doses of sunitinib in vitro and was synergistic with sunitinib in impairing tumor growth in vivo, indicating that these responses are triggered as prosurvival mechanisms that enable cells to tolerate the cytotoxic effects of sunitinib. Sunitinib 89-98 CREB regulated transcription coactivator 1 Mus musculus 23-29 27893461-4 2017 Inhibition of MCL-1 or mTORC1 signaling sensitized cells to clinically relevant doses of sunitinib in vitro and was synergistic with sunitinib in impairing tumor growth in vivo, indicating that these responses are triggered as prosurvival mechanisms that enable cells to tolerate the cytotoxic effects of sunitinib. Sunitinib 133-142 CREB regulated transcription coactivator 1 Mus musculus 23-29 27893461-4 2017 Inhibition of MCL-1 or mTORC1 signaling sensitized cells to clinically relevant doses of sunitinib in vitro and was synergistic with sunitinib in impairing tumor growth in vivo, indicating that these responses are triggered as prosurvival mechanisms that enable cells to tolerate the cytotoxic effects of sunitinib. Sunitinib 133-142 CREB regulated transcription coactivator 1 Mus musculus 23-29 27893461-5 2017 Furthermore, higher doses of sunitinib were cytotoxic, triggered a decline in MCL-1 levels, and inhibited mTORC1 signaling. Sunitinib 29-38 MCL1 apoptosis regulator, BCL2 family member Homo sapiens 78-83 27893461-5 2017 Furthermore, higher doses of sunitinib were cytotoxic, triggered a decline in MCL-1 levels, and inhibited mTORC1 signaling. Sunitinib 29-38 CREB regulated transcription coactivator 1 Mus musculus 106-112 27893461-6 2017 Mechanistically, we determined that sunitinib modulates MCL-1 stability by affecting its proteasomal degradation. Sunitinib 36-45 MCL1 apoptosis regulator, BCL2 family member Homo sapiens 56-61 27893461-7 2017 Dual modulation of MCL-1 stability at different dose ranges of sunitinib was due to differential effects on ERK and GSK3beta activity, and the latter also accounted for dual modulation of mTORC1 activity. Sunitinib 63-72 MCL1 apoptosis regulator, BCL2 family member Homo sapiens 19-24 27893461-7 2017 Dual modulation of MCL-1 stability at different dose ranges of sunitinib was due to differential effects on ERK and GSK3beta activity, and the latter also accounted for dual modulation of mTORC1 activity. Sunitinib 63-72 EPH receptor B2 Homo sapiens 108-111 27893461-7 2017 Dual modulation of MCL-1 stability at different dose ranges of sunitinib was due to differential effects on ERK and GSK3beta activity, and the latter also accounted for dual modulation of mTORC1 activity. Sunitinib 63-72 glycogen synthase kinase 3 beta Homo sapiens 116-124 27893461-8 2017 Finally, comparison of patient samples prior to and following sunitinib treatment suggested that increases in MCL-1 levels and mTORC1 activity correlate with resistance to sunitinib in patients. Sunitinib 62-71 MCL1 apoptosis regulator, BCL2 family member Homo sapiens 110-115 27893461-8 2017 Finally, comparison of patient samples prior to and following sunitinib treatment suggested that increases in MCL-1 levels and mTORC1 activity correlate with resistance to sunitinib in patients. Sunitinib 62-71 CREB regulated transcription coactivator 1 Mus musculus 127-133 27893461-8 2017 Finally, comparison of patient samples prior to and following sunitinib treatment suggested that increases in MCL-1 levels and mTORC1 activity correlate with resistance to sunitinib in patients. Sunitinib 172-181 MCL1 apoptosis regulator, BCL2 family member Homo sapiens 110-115 27893461-8 2017 Finally, comparison of patient samples prior to and following sunitinib treatment suggested that increases in MCL-1 levels and mTORC1 activity correlate with resistance to sunitinib in patients. Sunitinib 172-181 CREB regulated transcription coactivator 1 Mus musculus 127-133 27564227-7 2017 In conclusion, our data showed that the ABCB1 rs2032582 GG genotype was associated with individual adverse events" susceptibility among Japanese patients treated with sunitinib in routine clinical settings. Sunitinib 167-176 ATP binding cassette subfamily B member 1 Homo sapiens 40-45 28381801-9 2017 In conclusion, STAT3 polymorphism may be a novel risk factor for sunitinib-induced stomatitis in patients with mRCC. Sunitinib 65-74 signal transducer and activator of transcription 3 Homo sapiens 15-20 27737332-0 2017 Sunitinib in the therapy of malignant paragangliomas: report on the efficacy in a SDHB mutation carrier and review of the literature. Sunitinib 0-9 succinate dehydrogenase complex iron sulfur subunit B Homo sapiens 82-86 27737332-4 2017 Their therapy is palliative and represented by different options among which antiangiogenic drugs, like sunitinib, have been hypothesized to be effective especially in malignant SDHB mutated tumors. Sunitinib 104-113 succinate dehydrogenase complex iron sulfur subunit B Homo sapiens 178-182 27737332-5 2017 We report the effects of sunitinib therapy in a SDHB mutation carrier affected by a malignant sPGL. Sunitinib 25-34 succinate dehydrogenase complex iron sulfur subunit B Homo sapiens 48-52 28381801-0 2017 Association of Single Nucleotide Polymorphisms in STAT3, ABCB1, and ABCG2 with Stomatitis in Patients with Metastatic Renal Cell Carcinoma Treated with Sunitinib: A Retrospective Analysis in Japanese Patients. Sunitinib 152-161 signal transducer and activator of transcription 3 Homo sapiens 50-55 28381801-2 2017 Sunitinib is a substrate of P-glycoprotein (multidrug resistance (MDR)-1/ABCB1) and breast-cancer resistance protein (BCRP/ABCG2). Sunitinib 0-9 ATP binding cassette subfamily B member 1 Homo sapiens 44-72 27942928-1 2017 PURPOSE: Efficacy of targeted agents, such as everolimus and sunitinib, has been demonstrated in prospective trials on patients with gastroenteropancreatic neuroendocrine tumors (GEP-NETs). Sunitinib 61-70 granulin precursor Homo sapiens 179-182 28381801-2 2017 Sunitinib is a substrate of P-glycoprotein (multidrug resistance (MDR)-1/ABCB1) and breast-cancer resistance protein (BCRP/ABCG2). Sunitinib 0-9 ATP binding cassette subfamily B member 1 Homo sapiens 73-78 28381801-2 2017 Sunitinib is a substrate of P-glycoprotein (multidrug resistance (MDR)-1/ABCB1) and breast-cancer resistance protein (BCRP/ABCG2). Sunitinib 0-9 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 84-116 28381801-2 2017 Sunitinib is a substrate of P-glycoprotein (multidrug resistance (MDR)-1/ABCB1) and breast-cancer resistance protein (BCRP/ABCG2). Sunitinib 0-9 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 118-122 28381801-2 2017 Sunitinib is a substrate of P-glycoprotein (multidrug resistance (MDR)-1/ABCB1) and breast-cancer resistance protein (BCRP/ABCG2). Sunitinib 0-9 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 123-128 28381801-3 2017 In this retrospective study, we evaluated the association between sunitinib-induced stomatitis and STAT3, ABCB1, and ABCG2 polymorphisms in patients with metastatic renal cell carcinoma (mRCC). Sunitinib 66-75 signal transducer and activator of transcription 3 Homo sapiens 99-104 28381801-3 2017 In this retrospective study, we evaluated the association between sunitinib-induced stomatitis and STAT3, ABCB1, and ABCG2 polymorphisms in patients with metastatic renal cell carcinoma (mRCC). Sunitinib 66-75 ATP binding cassette subfamily B member 1 Homo sapiens 106-111 28381801-3 2017 In this retrospective study, we evaluated the association between sunitinib-induced stomatitis and STAT3, ABCB1, and ABCG2 polymorphisms in patients with metastatic renal cell carcinoma (mRCC). Sunitinib 66-75 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 117-122 27942928-10 2017 CONCLUSIONS: Both everolimus and sunitinib were effective in GEP-NET patients. Sunitinib 33-42 granulin precursor Homo sapiens 61-64 27942928-3 2017 METHODS: Clinical records of 44 patients with GEP-NET who were treated with everolimus or sunitinib between March 2007 and October 2014 were retrospectively reviewed. Sunitinib 90-99 granulin precursor Homo sapiens 46-49 29073615-0 2017 Mir-144-3p Promotes Cell Proliferation, Metastasis, Sunitinib Resistance in Clear Cell Renal Cell Carcinoma by Downregulating ARID1A. Sunitinib 52-61 microRNA 144 Mus musculus 0-7 29359059-5 2017 Four other cases of AIN reported along with inhibition of the vascular endothelial growth factor (VEGF) by either TKI (sunitinib and sorafenib) or antibodies (bevacizumab) suggest a possible class effect. Sunitinib 119-128 vascular endothelial growth factor A Homo sapiens 62-96 29359059-5 2017 Four other cases of AIN reported along with inhibition of the vascular endothelial growth factor (VEGF) by either TKI (sunitinib and sorafenib) or antibodies (bevacizumab) suggest a possible class effect. Sunitinib 119-128 vascular endothelial growth factor A Homo sapiens 98-102 28478450-4 2017 RESULTS: The results revealed that both the PDGF receptor-tyrosine kinase inhibitor imatinib and the multi-targeted VEGF and PDGF receptor inhibit or sunitinib malate reversed hypoxia-induced increases in right ventricular systolic pressure (RVSP), right ventricular function and thickening of the medial walls. Sunitinib 150-166 vascular endothelial growth factor A Rattus norvegicus 116-120 29073615-3 2017 This study aims to explore the roles of miR-144-3p in the invasion, migration and Sunitinib-resistance in ccRCC and to elucidate the underlying mechanisms. Sunitinib 82-91 microRNA 144 Mus musculus 40-47 29073615-12 2017 CONCLUSION: Higher miR-144-3p may enhance malignancy and resistance to Sunitinib in ccRCC by targeting ARID1A, the observations may uncover novel strategies of ccRCC treatment. Sunitinib 71-80 microRNA 144 Mus musculus 19-26 29073615-12 2017 CONCLUSION: Higher miR-144-3p may enhance malignancy and resistance to Sunitinib in ccRCC by targeting ARID1A, the observations may uncover novel strategies of ccRCC treatment. Sunitinib 71-80 AT rich interactive domain 1A (SWI-like) Mus musculus 103-109 28994095-12 2017 The immunoreactivities of Notch1, Jagged1, DLL-1 and VEGF in hepatocytes were significantly lower in the DM group when compared with the C, DM + S and C + S group treated with sunitinib. Sunitinib 176-185 notch 1 Mus musculus 26-32 27784207-11 2017 Specifically, in tumors resistant to single-agent sunitinib we detected a transcriptional response to hypoxia characterized by over-expression of several HIF1alpha target genes. Sunitinib 50-59 hypoxia inducible factor 1 subunit alpha Homo sapiens 154-163 28994095-12 2017 The immunoreactivities of Notch1, Jagged1, DLL-1 and VEGF in hepatocytes were significantly lower in the DM group when compared with the C, DM + S and C + S group treated with sunitinib. Sunitinib 176-185 jagged 1 Mus musculus 34-41 28994095-12 2017 The immunoreactivities of Notch1, Jagged1, DLL-1 and VEGF in hepatocytes were significantly lower in the DM group when compared with the C, DM + S and C + S group treated with sunitinib. Sunitinib 176-185 delta like canonical Notch ligand 1 Mus musculus 43-48 28994095-12 2017 The immunoreactivities of Notch1, Jagged1, DLL-1 and VEGF in hepatocytes were significantly lower in the DM group when compared with the C, DM + S and C + S group treated with sunitinib. Sunitinib 176-185 vascular endothelial growth factor A Mus musculus 53-57 28994095-13 2017 CONCLUSIONS: These results suggest that sunitinib effectively protects the liver from diabetes-induced damage through the inhibition of the Notch pathway. Sunitinib 40-49 notch 1 Mus musculus 140-145 28954991-0 2017 Antiangiogenic agent sunitinib induces epithelial to mesenchymal transition and accelerates motility of colorectal cancer cells. Sunitinib 21-30 collagen type XVIII alpha 1 chain Homo sapiens 0-20 28954991-4 2017 To elucidate a direct effect of VEGF-R-targeting drug (sunitinib), we established a human colorectal cancer cell model adapted to sunitinib. Sunitinib 55-64 kinase insert domain receptor Homo sapiens 32-38 28954991-6 2017 Consistent with the phenotype, the sunitinib-conditioned cells decreased the expression levels of E-cadherin (an epithelial marker), while significantly increased the levels of Slug and Zeb1 (mesenchymal markers). Sunitinib 35-44 cadherin 1 Homo sapiens 98-108 28954991-6 2017 Consistent with the phenotype, the sunitinib-conditioned cells decreased the expression levels of E-cadherin (an epithelial marker), while significantly increased the levels of Slug and Zeb1 (mesenchymal markers). Sunitinib 35-44 snail family transcriptional repressor 2 Homo sapiens 177-181 28954991-6 2017 Consistent with the phenotype, the sunitinib-conditioned cells decreased the expression levels of E-cadherin (an epithelial marker), while significantly increased the levels of Slug and Zeb1 (mesenchymal markers). Sunitinib 35-44 zinc finger E-box binding homeobox 1 Homo sapiens 186-190 28954991-7 2017 Expression profiles of VEGF-R in the sunitinib-conditioned cells showed that only neuropilin-1 (NRP1) expression was significantly increased among all VEGF-R tested. Sunitinib 37-46 kinase insert domain receptor Homo sapiens 23-29 28954991-7 2017 Expression profiles of VEGF-R in the sunitinib-conditioned cells showed that only neuropilin-1 (NRP1) expression was significantly increased among all VEGF-R tested. Sunitinib 37-46 neuropilin 1 Homo sapiens 82-94 28954991-7 2017 Expression profiles of VEGF-R in the sunitinib-conditioned cells showed that only neuropilin-1 (NRP1) expression was significantly increased among all VEGF-R tested. Sunitinib 37-46 neuropilin 1 Homo sapiens 96-100 28954991-7 2017 Expression profiles of VEGF-R in the sunitinib-conditioned cells showed that only neuropilin-1 (NRP1) expression was significantly increased among all VEGF-R tested. Sunitinib 37-46 kinase insert domain receptor Homo sapiens 151-157 28954991-8 2017 Blockade of NRP1 using its antagonist clearly repressed the migration activation in sunitinib-conditioned cells, but not in the control cells. Sunitinib 84-93 neuropilin 1 Homo sapiens 12-16 28478454-8 2017 RESULTS: VEGF (50 and 100 ng/ml) significantly augmented endothelial arginine transport in a time dependent manner, an effect which was prevented by Sunitinib (2 microM), a multi targeted receptor tyrosine kinase inhibitor. Sunitinib 149-158 vascular endothelial growth factor A Homo sapiens 9-13 28123542-0 2017 Influence of VEGFR single nucleotide polymorphisms on the efficacy of sunitinib therapy against renal cell carcinoma. Sunitinib 70-79 kinase insert domain receptor Homo sapiens 13-18 26810136-0 2017 Sunitinib-induced hypertension in CYP3A4 rs4646437 A-allele carriers with metastatic renal cell carcinoma. Sunitinib 0-9 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 34-40 26810136-1 2017 The single nucleotide polymorphism (SNP) rs4646437G>A in CYP3A4 was suggested to be related to sunitinib toxicity. Sunitinib 98-107 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 60-66 27840968-8 2017 Knockdown of ERK reduced sensitivity to sorafenib in both cell lines, knockdown of ERK and 4EBP1 reduced sensitivity to sunitinib in 769-P cells, and knockdown of 4EBP1 and p70 reduced sensitivity to everolimus in 786-O cells. Sunitinib 120-129 mitogen-activated protein kinase 1 Homo sapiens 83-96 27840968-8 2017 Knockdown of ERK reduced sensitivity to sorafenib in both cell lines, knockdown of ERK and 4EBP1 reduced sensitivity to sunitinib in 769-P cells, and knockdown of 4EBP1 and p70 reduced sensitivity to everolimus in 786-O cells. Sunitinib 120-129 eukaryotic translation initiation factor 4E binding protein 1 Homo sapiens 91-96 27840968-9 2017 High expression of phospho-ERK, -4EBP1 and -p70 correlated with better progression-free survival in patients treated with sorafenib, sunitinib and everolimus, respectively. Sunitinib 133-142 mitogen-activated protein kinase 1 Homo sapiens 27-30 27840968-9 2017 High expression of phospho-ERK, -4EBP1 and -p70 correlated with better progression-free survival in patients treated with sorafenib, sunitinib and everolimus, respectively. Sunitinib 133-142 eukaryotic translation initiation factor 4E binding protein 1 Homo sapiens 32-38 27840968-9 2017 High expression of phospho-ERK, -4EBP1 and -p70 correlated with better progression-free survival in patients treated with sorafenib, sunitinib and everolimus, respectively. Sunitinib 133-142 annexin A6 Homo sapiens 44-47 26810136-7 2017 In conclusion, hypertension is more likely to occur in A-allele carriers of the CYP3A4 rs4646437 variant in our cohort of mRCC patients treated with sunitinib. Sunitinib 149-158 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 80-86 28123542-1 2017 Single nucleotide polymorphisms (SNPs) of vascular endothelial growth factor receptor (VEGFR) may have effects on the MAPK/ERK/STAT3 signaling pathway, and the resulting phenotypes may influence the response to sunitinib-targeted therapy for renal cell carcinoma. Sunitinib 211-220 kinase insert domain receptor Homo sapiens 42-85 28123542-1 2017 Single nucleotide polymorphisms (SNPs) of vascular endothelial growth factor receptor (VEGFR) may have effects on the MAPK/ERK/STAT3 signaling pathway, and the resulting phenotypes may influence the response to sunitinib-targeted therapy for renal cell carcinoma. Sunitinib 211-220 kinase insert domain receptor Homo sapiens 87-92 28123542-1 2017 Single nucleotide polymorphisms (SNPs) of vascular endothelial growth factor receptor (VEGFR) may have effects on the MAPK/ERK/STAT3 signaling pathway, and the resulting phenotypes may influence the response to sunitinib-targeted therapy for renal cell carcinoma. Sunitinib 211-220 mitogen-activated protein kinase 1 Homo sapiens 123-126 28123542-1 2017 Single nucleotide polymorphisms (SNPs) of vascular endothelial growth factor receptor (VEGFR) may have effects on the MAPK/ERK/STAT3 signaling pathway, and the resulting phenotypes may influence the response to sunitinib-targeted therapy for renal cell carcinoma. Sunitinib 211-220 signal transducer and activator of transcription 3 Homo sapiens 127-132 28123542-14 2017 VEGFR SNPs are able to mediate the MAPK/ERK/STAT3 signaling pathway and therefore influence the effectiveness of sunitinib-targeted therapy, which makes them possible new therapeutic targets. Sunitinib 113-122 kinase insert domain receptor Homo sapiens 0-5 28123542-14 2017 VEGFR SNPs are able to mediate the MAPK/ERK/STAT3 signaling pathway and therefore influence the effectiveness of sunitinib-targeted therapy, which makes them possible new therapeutic targets. Sunitinib 113-122 mitogen-activated protein kinase 1 Homo sapiens 40-43 28123542-14 2017 VEGFR SNPs are able to mediate the MAPK/ERK/STAT3 signaling pathway and therefore influence the effectiveness of sunitinib-targeted therapy, which makes them possible new therapeutic targets. Sunitinib 113-122 signal transducer and activator of transcription 3 Homo sapiens 44-49 27974690-4 2016 The specific VHL CNL has not previously been associated with PNET, but has been reported in other tumors and has been associated with response to Sunitinib. Sunitinib 146-155 von Hippel-Lindau tumor suppressor Homo sapiens 13-16 28042343-3 2017 In the first mouse model, only sunitinib exhibited broad-spectrum antivascular and antitumor activities by simultaneously suppressing vascular endothelial growth factor receptor-2 (VEGFR2) and desmin expression, and by increasing intratumoral hypoxia and inhibiting both tumor growth and vascularisation significantly. Sunitinib 31-40 kinase insert domain protein receptor Mus musculus 134-179 28042343-3 2017 In the first mouse model, only sunitinib exhibited broad-spectrum antivascular and antitumor activities by simultaneously suppressing vascular endothelial growth factor receptor-2 (VEGFR2) and desmin expression, and by increasing intratumoral hypoxia and inhibiting both tumor growth and vascularisation significantly. Sunitinib 31-40 kinase insert domain protein receptor Mus musculus 181-187 27873490-7 2017 EGF activated epidermal growth factor receptor (EGFR) and triggered resistance to sunitinib, E7080, vandetanib, and sorafenib by transducing bypass survival signaling through ERK and AKT. Sunitinib 82-91 epidermal growth factor Homo sapiens 0-3 27974690-6 2016 To our knowledge, this is the first report of pulmonary PNET with CNL of VHL gene that benefits from Sunitinib treatment. Sunitinib 101-110 von Hippel-Lindau tumor suppressor Homo sapiens 73-76 28723497-9 2016 In the sunitinib cohort, the CYP3A4 rs464637 AG variant was associated with a lower risk of high-grade AEs (odds ratio 0.27, 95% confidence interval 0.08-0.88; p=0.03), but no SNPs were associated with hypertension. Sunitinib 7-16 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 29-35 28723497-11 2016 CONCLUSIONS: We observed an association between CYP3A4 polymorphisms and toxicity outcomes in mRCC patients treated with sunitinib, but not with everolimus or temsirolimus. Sunitinib 121-130 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 48-54 28723497-13 2016 PATIENT SUMMARY: We found that variants of CYP3A4, a gene involved in drug metabolism, are associated with sunitinib toxicity. Sunitinib 107-116 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 43-49 27921007-0 2016 Long-term Response to Nivolumab and Acute Renal Failure in a Patient with Metastatic Papillary Renal Cell Carcinoma and a PD-L1 Tumor Expression Increased with Sunitinib Therapy: A Case Report. Sunitinib 160-169 CD274 molecule Homo sapiens 122-127 27718781-1 2016 BACKGROUND: Sunitinib, a vascular endothelial growth factor pathway inhibitor, is an effective treatment for metastatic renal-cell carcinoma. Sunitinib 12-21 vascular endothelial growth factor A Homo sapiens 25-59 27417418-0 2016 Validation of VEGFR1 rs9582036 as predictive biomarker in metastatic clear-cell renal cell carcinoma patients treated with sunitinib. Sunitinib 123-132 fms related receptor tyrosine kinase 1 Homo sapiens 14-20 27417418-1 2016 OBJECTIVES: To validate vascular endothelial growth factor receptor-1 (VEGFR1) single nucleotide polymorphism (SNP) rs9582036 as a potential predictive biomarker in metastatic clear-cell renal cell carcinoma (m-ccRCC) patients treated with sunitinib. Sunitinib 240-249 fms related receptor tyrosine kinase 1 Homo sapiens 24-69 27417418-1 2016 OBJECTIVES: To validate vascular endothelial growth factor receptor-1 (VEGFR1) single nucleotide polymorphism (SNP) rs9582036 as a potential predictive biomarker in metastatic clear-cell renal cell carcinoma (m-ccRCC) patients treated with sunitinib. Sunitinib 240-249 fms related receptor tyrosine kinase 1 Homo sapiens 71-77 27417418-13 2016 CONCLUSION: VEGFR1 rs9582036 is a candidate predictive biomarker in m-ccRCC-patients treated with sunitinib. Sunitinib 98-107 fms related receptor tyrosine kinase 1 Homo sapiens 12-18 28008275-5 2016 GISTs with KIT mutations at exon 9 and the so-called wild-type GISTs seem to better respond to sunitinib. Sunitinib 95-104 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 11-14 28008275-6 2016 Nonetheless, further investigation is required to confirm these findings as well as to understand the mechanisms of sunitinib resistance such as the development of new KIT mutations or conformational changes in KIT receptor. Sunitinib 116-125 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 168-171 28008275-6 2016 Nonetheless, further investigation is required to confirm these findings as well as to understand the mechanisms of sunitinib resistance such as the development of new KIT mutations or conformational changes in KIT receptor. Sunitinib 116-125 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 211-214 28105174-11 2016 Thus, sunitinib confers radiosensitivity to esophageal cancer cells, which is associated with the downregulation of HIF-1alpha and VEGF expression. Sunitinib 6-15 hypoxia inducible factor 1 subunit alpha Homo sapiens 116-126 28105174-11 2016 Thus, sunitinib confers radiosensitivity to esophageal cancer cells, which is associated with the downregulation of HIF-1alpha and VEGF expression. Sunitinib 6-15 vascular endothelial growth factor A Homo sapiens 131-135 27459381-1 2016 Sunitinib and sorafenib are broad-spectrum tyrosine kinase inhibitors (TKI) targeting, for example, VEGF1-3, PDGFRb, RET, FLT3, CD117 (c-KIT) and CSF-1R cell membrane receptors thus suppressing tumor angiogenesis and cancer cell growth. Sunitinib 0-9 platelet derived growth factor receptor, beta polypeptide Mus musculus 109-115 27459381-1 2016 Sunitinib and sorafenib are broad-spectrum tyrosine kinase inhibitors (TKI) targeting, for example, VEGF1-3, PDGFRb, RET, FLT3, CD117 (c-KIT) and CSF-1R cell membrane receptors thus suppressing tumor angiogenesis and cancer cell growth. Sunitinib 0-9 ret proto-oncogene Mus musculus 117-120 27459381-1 2016 Sunitinib and sorafenib are broad-spectrum tyrosine kinase inhibitors (TKI) targeting, for example, VEGF1-3, PDGFRb, RET, FLT3, CD117 (c-KIT) and CSF-1R cell membrane receptors thus suppressing tumor angiogenesis and cancer cell growth. Sunitinib 0-9 FMS-like tyrosine kinase 3 Mus musculus 122-126 27459381-1 2016 Sunitinib and sorafenib are broad-spectrum tyrosine kinase inhibitors (TKI) targeting, for example, VEGF1-3, PDGFRb, RET, FLT3, CD117 (c-KIT) and CSF-1R cell membrane receptors thus suppressing tumor angiogenesis and cancer cell growth. Sunitinib 0-9 KIT proto-oncogene receptor tyrosine kinase Mus musculus 128-133 27459381-1 2016 Sunitinib and sorafenib are broad-spectrum tyrosine kinase inhibitors (TKI) targeting, for example, VEGF1-3, PDGFRb, RET, FLT3, CD117 (c-KIT) and CSF-1R cell membrane receptors thus suppressing tumor angiogenesis and cancer cell growth. Sunitinib 0-9 KIT proto-oncogene receptor tyrosine kinase Mus musculus 135-140 27633584-8 2016 Immunohistochemistry analyses found that both sunitinib (60 mg/kg) and HM-3 (3 and 48 mg/kg) decreased microvascular density and increased percent of HIF-1alpha and VEGF expressing cells. Sunitinib 46-55 hypoxia inducible factor 1 subunit alpha Homo sapiens 150-160 27841748-5 2016 Suppressing RET activity using Sunitinib, a clinically-approved tyrosine kinase inhibitor, rescued differentiation in both DUX4-expressing murine myoblasts and in FSHD patient-derived myoblasts. Sunitinib 31-40 ret proto-oncogene Mus musculus 12-15 27841748-5 2016 Suppressing RET activity using Sunitinib, a clinically-approved tyrosine kinase inhibitor, rescued differentiation in both DUX4-expressing murine myoblasts and in FSHD patient-derived myoblasts. Sunitinib 31-40 double homeobox Mus musculus 123-127 27841748-8 2016 Rescue of DUX4-induced pathology by Sunitinib highlights the therapeutic potential of tyrosine kinase inhibitors for treatment of FSHD. Sunitinib 36-45 double homeobox Mus musculus 10-14 27706901-5 2016 These data, together with molecular docking simulations, revealed distinct differences in the impact of structural modifications on drug binding to the enzymes: whereas the catalytic pocket of the epidermal growth factor receptor (EGFR) accepted all new erlotinib derivatives, the vascular endothelial growth factor receptor (VEGFR)-inhibitory potential in the case of the sunitinib prodrugs was dramatically diminished by derivatization. Sunitinib 373-382 epidermal growth factor receptor Homo sapiens 197-229 27706901-5 2016 These data, together with molecular docking simulations, revealed distinct differences in the impact of structural modifications on drug binding to the enzymes: whereas the catalytic pocket of the epidermal growth factor receptor (EGFR) accepted all new erlotinib derivatives, the vascular endothelial growth factor receptor (VEGFR)-inhibitory potential in the case of the sunitinib prodrugs was dramatically diminished by derivatization. Sunitinib 373-382 epidermal growth factor receptor Homo sapiens 231-235 27793938-0 2016 FoxO3a is Essential for the Antiproliferative and Apoptogenic Effects of Sunitinib in MDA-MB231 Cell Line. Sunitinib 73-82 forkhead box O3 Homo sapiens 0-6 27930642-4 2016 In a phase 3 adjuvant therapy trial (S-TRAC), sunitinib increased median disease-free survival in patients with clear cell RCC who were at very high risk. Sunitinib 46-55 T cell receptor alpha constant Homo sapiens 39-43 27822356-5 2016 However, several molecularly targeted drugs have been proposed as potentially beneficial, including tyrosine kinase inhibitors (TKIs) directed at vascular endothelial growth factor receptor (VEGFR), like pazopanib and sunitinib. Sunitinib 218-227 kinase insert domain receptor Homo sapiens 191-196 28133332-6 2016 Mutational analysis revealed a PDGFRA mutation in exon 18, which causes resistance to both imatinib and sunitinib. Sunitinib 104-113 platelet derived growth factor receptor alpha Homo sapiens 31-37 27633584-8 2016 Immunohistochemistry analyses found that both sunitinib (60 mg/kg) and HM-3 (3 and 48 mg/kg) decreased microvascular density and increased percent of HIF-1alpha and VEGF expressing cells. Sunitinib 46-55 vascular endothelial growth factor A Homo sapiens 165-169 27495788-1 2016 PURPOSE: Pharmacokinetic interaction of sunitinib with diclofenac, paracetamol, mefenamic acid and ibuprofen was evaluated due to their P450 mediated metabolism and OATP1B1, OATP1B3, ABCB1, ABCG2 transporters overlapping features. Sunitinib 40-49 ATP-binding cassette, sub-family B (MDR/TAP), member 1B Mus musculus 183-188 27560553-0 2016 MEK inhibition abrogates sunitinib resistance in a renal cell carcinoma patient-derived xenograft model. Sunitinib 25-34 mitogen-activated protein kinase kinase 7 Homo sapiens 0-3 26416414-7 2016 No difference was found in patients treated with sunitinib (P = 0.370).A second line with sunitinib was associated with an improved TTP in KIT exon 11 mutated patients progressing on imatinib 400 mg/die. Sunitinib 90-99 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 139-142 27920695-5 2016 We report two individuals with metastatic ASPS who obtained disease stabilization from sunitinib lasting over a year. Sunitinib 87-96 ASPSCR1 tether for SLC2A4, UBX domain containing Homo sapiens 42-46 27495788-1 2016 PURPOSE: Pharmacokinetic interaction of sunitinib with diclofenac, paracetamol, mefenamic acid and ibuprofen was evaluated due to their P450 mediated metabolism and OATP1B1, OATP1B3, ABCB1, ABCG2 transporters overlapping features. Sunitinib 40-49 ATP binding cassette subfamily G member 2 (Junior blood group) Mus musculus 190-195 27525572-9 2016 Also, sunitinib dose dependently reduced collagen- and ADP-induced aggregation, collagen-dependent thrombus formation and collagen-induced secretion of platelet-derived growth factor and beta-thromboglobulin. Sunitinib 6-15 pro-platelet basic protein Homo sapiens 187-207 27450971-2 2016 The first-generation FLT3 inhibitors were developed several years ago and include midostaurin, lestaurtinib, sunitinib, and sorafenib. Sunitinib 109-118 fms related receptor tyrosine kinase 3 Homo sapiens 21-25 27525572-13 2016 CONCLUSIONS: Sunitinib uptake by platelets inhibits collagen receptor-induced aggregation and thrombus formation via reduction of protein tyrosine phosphorylation and alpha-granule secretion. Sunitinib 13-22 integrin subunit alpha 2 Homo sapiens 52-69 27432152-8 2016 Tumorsphere formation is one of the indicators of aggressiveness in malignant cancers, which was significantly inhibited in IMR-32 cells after L1-CAM KD or Sutent treatment, however, no synergistic effect was observed with dual treatments, rather L1-CAM KD alone showed a greater inhibition on tumorsphere formation compared to Sutent treatment alone. Sunitinib 328-334 L1 cell adhesion molecule Homo sapiens 143-149 26755746-8 2016 Of interest, VEGF inhibition with drugs like sunitinib, applied in cancer patients, results in a PE-like syndrome, characterized by hypertension, proteinuria and renal toxicity. Sunitinib 45-54 vascular endothelial growth factor A Homo sapiens 13-17 26755746-9 2016 Both in cancer patients treated with sunitinib and in pregnant women with PE, significant rises in endothelin-1 occur. Sunitinib 37-46 endothelin 1 Homo sapiens 99-111 27733829-12 2016 The sequence Sunitinib-Axitinib was well-tolerated without worsening in side effects, with a median OS of 34.7 months (95% CI 18.4-51.0 months). Sunitinib 13-22 Moloney sarcoma oncogene Mus musculus 100-102 27686636-3 2016 Results: The levels of CD3+ , CD8+ T lymphocyte, NK cell, B lymphocyte were significantly increased after the therapy of Sunitinib for 1 cycle(I-J was 212+-22, 163+-18, 59+-12, 13.8+-1.4, respectively, all P<0.05). Sunitinib 121-130 CD8a molecule Homo sapiens 30-33 27686636-4 2016 The levels of CD3+ , CD8+ T lymphocyte, NK cell, B lymphocyte were significantly increased after the therapy of Sunitinib for 2 cycles(I-J was 362+-43, 299+-28, 91+-19, 28.1+-3.9, respectively, all P<0.05), while the level of CD4+ lymphocyte was decreased, but no significant difference(F=0.873, P>0.05). Sunitinib 112-121 CD8a molecule Homo sapiens 21-24 27686636-4 2016 The levels of CD3+ , CD8+ T lymphocyte, NK cell, B lymphocyte were significantly increased after the therapy of Sunitinib for 2 cycles(I-J was 362+-43, 299+-28, 91+-19, 28.1+-3.9, respectively, all P<0.05), while the level of CD4+ lymphocyte was decreased, but no significant difference(F=0.873, P>0.05). Sunitinib 112-121 CD4 molecule Homo sapiens 229-232 27686636-5 2016 CD4+ /CD8+ was significantly decreased after the therapy of Sunitinib for 1 cycle, and it went on decreasing after the therapy of Sunitinib for 2 cycles(I-J was -0.31+-0.03, -0.44+-0.04, respectively, all P<0.05). Sunitinib 60-69 CD4 molecule Homo sapiens 0-3 27686636-5 2016 CD4+ /CD8+ was significantly decreased after the therapy of Sunitinib for 1 cycle, and it went on decreasing after the therapy of Sunitinib for 2 cycles(I-J was -0.31+-0.03, -0.44+-0.04, respectively, all P<0.05). Sunitinib 60-69 CD8a molecule Homo sapiens 6-9 27686636-5 2016 CD4+ /CD8+ was significantly decreased after the therapy of Sunitinib for 1 cycle, and it went on decreasing after the therapy of Sunitinib for 2 cycles(I-J was -0.31+-0.03, -0.44+-0.04, respectively, all P<0.05). Sunitinib 130-139 CD4 molecule Homo sapiens 0-3 27506945-8 2016 Compared with HR-negative tumor, HR-positive tumor had significantly better response to sunitinib (p = 0.035). Sunitinib 88-97 nuclear receptor subfamily 4 group A member 1 Homo sapiens 33-35 27556517-0 2016 Adrenomedullin blockade suppresses sunitinib-resistant renal cell carcinoma growth by targeting the ERK/MAPK pathway. Sunitinib 35-44 mitogen-activated protein kinase 1 Mus musculus 100-103 27556517-0 2016 Adrenomedullin blockade suppresses sunitinib-resistant renal cell carcinoma growth by targeting the ERK/MAPK pathway. Sunitinib 35-44 mitogen-activated protein kinase 1 Mus musculus 104-108 27556517-3 2016 And adrenomedullin expression was increased in sunitinib-resistant tumor xenografts, accompanied by upregulation of phospho-ERK levels. Sunitinib 47-56 mitogen-activated protein kinase 1 Mus musculus 124-127 27164264-9 2016 Sensitive markers for sunitinib therapy include KDR positivity and early-developed treatment-induced hypertension. Sunitinib 22-31 kinase insert domain receptor Homo sapiens 48-51 27164264-10 2016 Resistance factors for sunitinib include VEGF positivity, CAIX positivity and squamous cell carcinoma pathology type. Sunitinib 23-32 carbonic anhydrase 9 Homo sapiens 58-62 27487138-8 2016 Ubiquitin-like with PHD and ring finger domains 1 (UHRF1) was directly suppressed by miR-101 in RCC cells, and overexpression of UHRF1 was confirmed in sunitinib-treated RCC tissues. Sunitinib 152-161 ubiquitin like with PHD and ring finger domains 1 Homo sapiens 0-49 27487138-8 2016 Ubiquitin-like with PHD and ring finger domains 1 (UHRF1) was directly suppressed by miR-101 in RCC cells, and overexpression of UHRF1 was confirmed in sunitinib-treated RCC tissues. Sunitinib 152-161 ubiquitin like with PHD and ring finger domains 1 Homo sapiens 51-56 27487138-8 2016 Ubiquitin-like with PHD and ring finger domains 1 (UHRF1) was directly suppressed by miR-101 in RCC cells, and overexpression of UHRF1 was confirmed in sunitinib-treated RCC tissues. Sunitinib 152-161 ubiquitin like with PHD and ring finger domains 1 Homo sapiens 129-134 27487138-10 2016 Our findings showed that antitumor miR-101- mediated UHRF1 pathways may be suppressed by sunitinib treatment. Sunitinib 89-98 ubiquitin like with PHD and ring finger domains 1 Homo sapiens 53-58 27288242-0 2016 The tyrosine kinase inhibitor, sunitinib malate, induces cognitive impairment in vivo via dysregulating VEGFR signaling, apoptotic and autophagic machineries. Sunitinib 31-47 kinase insert domain receptor Homo sapiens 104-109 26597115-1 2016 PURPOSE: This study aimed to clarify the molecular mechanism mediating the cytotoxicity of axitinib, a selective inhibitor of the vascular endothelial growth factor receptor (VEGFR), in sunitinib-resistant renal cell carcinoma (RCC). Sunitinib 186-195 kinase insert domain receptor Homo sapiens 130-173 26597115-1 2016 PURPOSE: This study aimed to clarify the molecular mechanism mediating the cytotoxicity of axitinib, a selective inhibitor of the vascular endothelial growth factor receptor (VEGFR), in sunitinib-resistant renal cell carcinoma (RCC). Sunitinib 186-195 kinase insert domain receptor Homo sapiens 175-180 27485537-0 2016 BSA and ABCB1 polymorphism affect the pharmacokinetics of sunitinib and its active metabolite in Asian mRCC patients receiving an attenuated sunitinib dosing regimen. Sunitinib 58-67 ATP binding cassette subfamily B member 1 Homo sapiens 8-13 27485537-7 2016 RESULTS: Sunitinib population means for CL/F and V d /F central were 13.8 L/h and 1720 L, respectively. Sunitinib 9-18 CLQTL1 Homo sapiens 40-52 27485537-9 2016 Body surface area (BSA) and ABCB1 polymorphism significantly influenced the CL/F variability of sunitinib: CL/F parent = 13.8 x exp((BSA - 1.75) x 2.08 + (ABCB1 genotype - 0.67) x 0.61), ABCB1-0: wild genotype, 1: mutant genotype. Sunitinib 96-105 ATP binding cassette subfamily B member 1 Homo sapiens 28-33 27485537-9 2016 Body surface area (BSA) and ABCB1 polymorphism significantly influenced the CL/F variability of sunitinib: CL/F parent = 13.8 x exp((BSA - 1.75) x 2.08 + (ABCB1 genotype - 0.67) x 0.61), ABCB1-0: wild genotype, 1: mutant genotype. Sunitinib 96-105 ATP binding cassette subfamily B member 1 Homo sapiens 155-160 27485537-9 2016 Body surface area (BSA) and ABCB1 polymorphism significantly influenced the CL/F variability of sunitinib: CL/F parent = 13.8 x exp((BSA - 1.75) x 2.08 + (ABCB1 genotype - 0.67) x 0.61), ABCB1-0: wild genotype, 1: mutant genotype. Sunitinib 96-105 ATP binding cassette subfamily B member 1 Homo sapiens 155-160 27485537-10 2016 The effect size of ABCB1 mutant genotype and BSA greater than 1.75 m(2) in relation to sunitinib clearance was 31.14 % (p = 0.006) and 22.11 % (p = 0.011), respectively, relative to the reference group. Sunitinib 87-96 ATP binding cassette subfamily B member 1 Homo sapiens 19-24 27485537-11 2016 CONCLUSIONS: Adjusting doses of sunitinib according to BSA and ABCB1 polymorphism in Asian mRCC patients may be recommended for sufficient attainment of a target TTL of sunitinib and its metabolite. Sunitinib 32-41 ATP binding cassette subfamily B member 1 Homo sapiens 63-68 27288242-11 2016 Besides, sunitinib boosted cortical and hippocampal p53 and executioner caspase-3 and decreased nuclear factor kappa B and Bcl-2 levels promoting apoptotic cell death. Sunitinib 9-18 tumor protein p53 Homo sapiens 52-55 27288242-11 2016 Besides, sunitinib boosted cortical and hippocampal p53 and executioner caspase-3 and decreased nuclear factor kappa B and Bcl-2 levels promoting apoptotic cell death. Sunitinib 9-18 caspase 3 Homo sapiens 72-81 27288242-11 2016 Besides, sunitinib boosted cortical and hippocampal p53 and executioner caspase-3 and decreased nuclear factor kappa B and Bcl-2 levels promoting apoptotic cell death. Sunitinib 9-18 BCL2 apoptosis regulator Homo sapiens 123-128 27288242-13 2016 To our knowledge, this is the first study to provide molecular insights into sunitinib-induced chemofog where impeded VEGFR2 signaling and autophagic and hyperactivated apoptotic machineries act in neurodegenerative concert. Sunitinib 77-86 kinase insert domain receptor Homo sapiens 118-124 27485825-1 2016 The von Hippel-Lindau tumor suppressor (VHL; E3 ubiquitin ligase gene) is frequently mutated or undetectable in clear cell renal cell carcinoma (CCRCC), and therefore these tumors are highly resistant to chemotherapeutic agents, including adriamycin (ADM) and sunitinib. Sunitinib 260-269 von Hippel-Lindau tumor suppressor Homo sapiens 40-43 27602218-0 2016 GLI2 expression levels in radical nephrectomy specimens as a predictor of disease progression in patients with metastatic clear cell renal cell carcinoma following treatment with sunitinib. Sunitinib 179-188 GLI family zinc finger 2 Homo sapiens 0-4 27485825-1 2016 The von Hippel-Lindau tumor suppressor (VHL; E3 ubiquitin ligase gene) is frequently mutated or undetectable in clear cell renal cell carcinoma (CCRCC), and therefore these tumors are highly resistant to chemotherapeutic agents, including adriamycin (ADM) and sunitinib. Sunitinib 260-269 Cbl proto-oncogene like 2 Homo sapiens 45-64 27485825-5 2016 The expression of VHL in the MZ1257 cell line sensitized these cells to ADM and sunitinib, and a knockdown of VHL in the ACHN cells increased their chemoresistance. Sunitinib 80-89 von Hippel-Lindau tumor suppressor Homo sapiens 18-21 27602218-6 2016 Furthermore, treatment with sunitinib resulted in a marked inhibition of GLI2 expression in the parental human RCC ACHN cell line, but not in ACHN cells with acquired resistance to sunitinib. Sunitinib 28-37 GLI family zinc finger 2 Homo sapiens 73-77 27602218-7 2016 These findings suggested that GLI2 may be involved in the acquisition of resistance to sunitinib in RCC; thus, it may be useful to consider the expression levels of GLI2 in addition to conventional prognostic parameters when selecting m-ccRCC patients likely to benefit from treatment with sunitinib. Sunitinib 87-96 GLI family zinc finger 2 Homo sapiens 30-34 27485825-9 2016 Further analysis indicated that co-expression of VHL and P53 inhibited cell proliferation by completely inhibiting the cell cycle at the G0/G1 phase, and promoted apoptosis following treatment with ADM or sunitinib. Sunitinib 205-214 von Hippel-Lindau tumor suppressor Homo sapiens 49-52 27485825-9 2016 Further analysis indicated that co-expression of VHL and P53 inhibited cell proliferation by completely inhibiting the cell cycle at the G0/G1 phase, and promoted apoptosis following treatment with ADM or sunitinib. Sunitinib 205-214 tumor protein p53 Homo sapiens 57-60 29263493-0 2016 [Sunitinib induces autophagy via suppressing Akt/mTOR pathway in renal cell carcinoma]. Sunitinib 1-10 AKT serine/threonine kinase 1 Homo sapiens 45-48 27235791-6 2016 Regarding the studied antifibrotics, gene levels of HSP47 and PCOL1A1 could be down-regulated with sunitinib and valproic acid, while PCOL1A1 expression was reduced following treatment with rosmarinic acid, tetrandrine and pirfenidone. Sunitinib 99-108 serpin family H member 1 Homo sapiens 52-57 27297795-6 2016 Examination of the underlying mechanisms indicated that venetoclax resistance may be induced by microenvironmental signals that upregulate antiapoptotic Bcl-xl, Mcl-1, and A1, which can be counteracted more efficiently by sunitinib than by ibrutinib or idelalisib. Sunitinib 222-231 MCL1 apoptosis regulator, BCL2 family member Homo sapiens 161-166 27580750-4 2016 Discontinuation of an anti-VEGF antibody-based drug and sunitinib markedly promotes liver metastasis. Sunitinib 56-65 vascular endothelial growth factor A Homo sapiens 27-31 29263493-0 2016 [Sunitinib induces autophagy via suppressing Akt/mTOR pathway in renal cell carcinoma]. Sunitinib 1-10 mechanistic target of rapamycin kinase Homo sapiens 49-53 29263493-14 2016 The rapamycin (mTOR inhibitor) or knocking down Akt subunits could change the sunitinib-induced LC3 -II accumulation, whereas overexpression of Akt subunits decreased the autophagic flux, indicating that Akt/mTOR was the target of sunitinib in autophagy. Sunitinib 78-87 mechanistic target of rapamycin kinase Homo sapiens 208-212 29263493-10 2016 And the nuclear-to-cytosol translocation as well as aggregation of LC3-II was presented after sunitinib treatment by the fluorescence microscope, which was the proof of the enhanced autophagy. Sunitinib 94-103 microtubule associated protein 1 light chain 3 alpha Homo sapiens 67-70 29263493-14 2016 The rapamycin (mTOR inhibitor) or knocking down Akt subunits could change the sunitinib-induced LC3 -II accumulation, whereas overexpression of Akt subunits decreased the autophagic flux, indicating that Akt/mTOR was the target of sunitinib in autophagy. Sunitinib 231-240 mechanistic target of rapamycin kinase Homo sapiens 208-212 29263493-15 2016 CONCLUSION: Sunitinib induced autophagy via suppressing Akt/mTOR pathway, and the autophagy was involved in apopotosis. Sunitinib 12-21 AKT serine/threonine kinase 1 Homo sapiens 56-59 29263493-11 2016 According to the immunoblotting, sunitinib was able to increase the accumulation of LC3-II . Sunitinib 33-42 microtubule associated protein 1 light chain 3 alpha Homo sapiens 84-87 29263493-15 2016 CONCLUSION: Sunitinib induced autophagy via suppressing Akt/mTOR pathway, and the autophagy was involved in apopotosis. Sunitinib 12-21 mechanistic target of rapamycin kinase Homo sapiens 60-64 29263493-12 2016 At the same time, the result of sunitinib combined with chloroquine, a drug which blocked the fusion of autophagosomes and lysosomes, demonstrated that the increasing amount of LC3-II was due to the enhanced autophagy flux by sunitinib treatment in ACHN cells. Sunitinib 32-41 microtubule associated protein 1 light chain 3 alpha Homo sapiens 177-180 29263494-11 2016 CONCLUSION: MEK/ERK signaling pathways may play an important role in the metastasis and the resistance of sunitinib in RCC patients with bone metastasis. Sunitinib 106-115 mitogen-activated protein kinase kinase 7 Homo sapiens 12-15 29263493-12 2016 At the same time, the result of sunitinib combined with chloroquine, a drug which blocked the fusion of autophagosomes and lysosomes, demonstrated that the increasing amount of LC3-II was due to the enhanced autophagy flux by sunitinib treatment in ACHN cells. Sunitinib 226-235 microtubule associated protein 1 light chain 3 alpha Homo sapiens 177-180 29263494-11 2016 CONCLUSION: MEK/ERK signaling pathways may play an important role in the metastasis and the resistance of sunitinib in RCC patients with bone metastasis. Sunitinib 106-115 mitogen-activated protein kinase 1 Homo sapiens 16-19 29263493-14 2016 The rapamycin (mTOR inhibitor) or knocking down Akt subunits could change the sunitinib-induced LC3 -II accumulation, whereas overexpression of Akt subunits decreased the autophagic flux, indicating that Akt/mTOR was the target of sunitinib in autophagy. Sunitinib 78-87 mechanistic target of rapamycin kinase Homo sapiens 15-19 29263493-14 2016 The rapamycin (mTOR inhibitor) or knocking down Akt subunits could change the sunitinib-induced LC3 -II accumulation, whereas overexpression of Akt subunits decreased the autophagic flux, indicating that Akt/mTOR was the target of sunitinib in autophagy. Sunitinib 78-87 AKT serine/threonine kinase 1 Homo sapiens 48-51 29263493-14 2016 The rapamycin (mTOR inhibitor) or knocking down Akt subunits could change the sunitinib-induced LC3 -II accumulation, whereas overexpression of Akt subunits decreased the autophagic flux, indicating that Akt/mTOR was the target of sunitinib in autophagy. Sunitinib 78-87 microtubule associated protein 1 light chain 3 alpha Homo sapiens 96-99 27016208-0 2016 Schedule-dependent cytotoxicity of sunitinib and TRAIL in human non-small cell lung cancer cells with or without EGFR and KRAS mutations. Sunitinib 35-44 epidermal growth factor receptor Homo sapiens 113-117 27046396-0 2016 Sunitinib, a Clinically Used Anticancer Drug, Is a Potent AChE Inhibitor and Attenuates Cognitive Impairments in Mice. Sunitinib 0-9 acetylcholinesterase Mus musculus 58-62 27046396-2 2016 In this study, we found for the first time that sunitinib inhibits acetylcholinesterase (AChE) at submicromolar concentrations in vitro. Sunitinib 48-57 acetylcholinesterase Mus musculus 67-87 27046396-2 2016 In this study, we found for the first time that sunitinib inhibits acetylcholinesterase (AChE) at submicromolar concentrations in vitro. Sunitinib 48-57 acetylcholinesterase Mus musculus 89-93 27046396-3 2016 In addition, sunitinib dramatically decreased the hippocampal and cortical activity of AChE in a time-dependent manner in mice. Sunitinib 13-22 acetylcholinesterase Mus musculus 87-91 27046396-4 2016 Molecular docking analysis further demonstrates that sunitinib might interact with both the catalytic anion and peripheral anionic sites within AChE, which is in accordance with enzymatic activity results showing that sunitinib inhibits AChE in a mixed pattern. Sunitinib 53-62 acetylcholinesterase Mus musculus 144-148 27046396-4 2016 Molecular docking analysis further demonstrates that sunitinib might interact with both the catalytic anion and peripheral anionic sites within AChE, which is in accordance with enzymatic activity results showing that sunitinib inhibits AChE in a mixed pattern. Sunitinib 53-62 acetylcholinesterase Mus musculus 237-241 27046396-4 2016 Molecular docking analysis further demonstrates that sunitinib might interact with both the catalytic anion and peripheral anionic sites within AChE, which is in accordance with enzymatic activity results showing that sunitinib inhibits AChE in a mixed pattern. Sunitinib 218-227 acetylcholinesterase Mus musculus 144-148 27046396-4 2016 Molecular docking analysis further demonstrates that sunitinib might interact with both the catalytic anion and peripheral anionic sites within AChE, which is in accordance with enzymatic activity results showing that sunitinib inhibits AChE in a mixed pattern. Sunitinib 218-227 acetylcholinesterase Mus musculus 237-241 27046396-6 2016 Surprisingly, sunitinib could attenuate cognitive impairments to a similar extent as donepezil, a marketed AChE inhibitor used for the treatment of Alzheimer"s disease. Sunitinib 14-23 acetylcholinesterase Mus musculus 107-111 27046396-7 2016 In summary, our results have shown that sunitinib could potently inhibit AChE and attenuate cognitive impairments in mice. Sunitinib 40-49 acetylcholinesterase Mus musculus 73-77 27488093-6 2016 Namely high HIF-2alpha, CD31, and CAIX expression levels along with low VEGFR1 and PDGFRB expression levels improved the benefit of sunitinib treatment compared with sorafenib treatment. Sunitinib 132-141 carbonic anhydrase 9 Homo sapiens 34-38 27488093-6 2016 Namely high HIF-2alpha, CD31, and CAIX expression levels along with low VEGFR1 and PDGFRB expression levels improved the benefit of sunitinib treatment compared with sorafenib treatment. Sunitinib 132-141 fms related receptor tyrosine kinase 1 Homo sapiens 72-78 27488093-6 2016 Namely high HIF-2alpha, CD31, and CAIX expression levels along with low VEGFR1 and PDGFRB expression levels improved the benefit of sunitinib treatment compared with sorafenib treatment. Sunitinib 132-141 platelet derived growth factor receptor beta Homo sapiens 83-89 27488093-5 2016 We found that patients with high HIF-2alpha, CD31 expression showed greater relative PFS and OS benefit and patients with high CAIX expression presented greater relative OS benefit from sunitinib than from sorafenib, patients with high VEGFR1 or PDGFRB expression levels exhibited worse relative PFS benefit from sunitinib than from sorafenib. Sunitinib 186-195 carbonic anhydrase 9 Homo sapiens 127-131 27488093-6 2016 Namely high HIF-2alpha, CD31, and CAIX expression levels along with low VEGFR1 and PDGFRB expression levels improved the benefit of sunitinib treatment compared with sorafenib treatment. Sunitinib 132-141 endothelial PAS domain protein 1 Homo sapiens 12-22 27488093-6 2016 Namely high HIF-2alpha, CD31, and CAIX expression levels along with low VEGFR1 and PDGFRB expression levels improved the benefit of sunitinib treatment compared with sorafenib treatment. Sunitinib 132-141 platelet and endothelial cell adhesion molecule 1 Homo sapiens 24-28 27016208-7 2016 We found that sunitinib enhances TRAIL-induced G0/G1-phase cell cycle arrest and blocks TRAIL-triggered activation of AKT as the underlying mechanism. Sunitinib 14-23 TNF superfamily member 10 Homo sapiens 33-38 27016208-7 2016 We found that sunitinib enhances TRAIL-induced G0/G1-phase cell cycle arrest and blocks TRAIL-triggered activation of AKT as the underlying mechanism. Sunitinib 14-23 TNF superfamily member 10 Homo sapiens 88-93 27016208-7 2016 We found that sunitinib enhances TRAIL-induced G0/G1-phase cell cycle arrest and blocks TRAIL-triggered activation of AKT as the underlying mechanism. Sunitinib 14-23 AKT serine/threonine kinase 1 Homo sapiens 118-121 27016208-8 2016 In contrast, we observed antagonistic effects when sunitinib was administered after TRAIL to the cell lines tested. Sunitinib 51-60 TNF superfamily member 10 Homo sapiens 84-89 27016208-10 2016 CONCLUSION: From our data we conclude that administration of sunitinib followed by TRAIL, as well as a simultaneous administration of both agents, serve as favorable treatment regimens for NSCLC-derived cells, irrespective of their EGFR and/or KRAS mutation status. Sunitinib 61-70 KRAS proto-oncogene, GTPase Homo sapiens 244-248 27374084-4 2016 We showed that Ela-myc pancreatic tumors express PDGFR and VEGFR in blood vessels and epithelial cells, rendering these tumors sensitive to sunitinib by more than only its anti-angiogenic activity. Sunitinib 140-149 platelet derived growth factor receptor, beta polypeptide Mus musculus 49-54 27123883-7 2016 Potential activity of VEGF (vascular endothelial growth factor) inhibitors such as sunitinib, valatinib, and bevacizumab is suggested in small non-controlled studies and requires validation in randomized trials. Sunitinib 83-92 vascular endothelial growth factor A Homo sapiens 22-26 27123883-7 2016 Potential activity of VEGF (vascular endothelial growth factor) inhibitors such as sunitinib, valatinib, and bevacizumab is suggested in small non-controlled studies and requires validation in randomized trials. Sunitinib 83-92 vascular endothelial growth factor A Homo sapiens 28-62 27439438-12 2016 Moreover, sunitinib reduced the degree of phosphorylation of serine residues on Smad2/3 that was induced by TGF-beta in HBEs. Sunitinib 10-19 SMAD family member 2 Mus musculus 80-87 27374084-5 2016 However, sunitinib treatment of Ela-myc mice with either early or advanced tumor progression had no impact on either survival or tumor burden. Sunitinib 9-18 apelin receptor early endogenous ligand Mus musculus 32-35 27374084-7 2016 In stark contrast, in vitro sunitinib treatment of Ela-myc- derived cell lines showed high sensitivity to the drug, with increased apoptosis and reduced proliferation. Sunitinib 28-37 apelin receptor early endogenous ligand Mus musculus 51-54 27304187-8 2016 Finally, we also found that PDGFRB is one of the components of a complex that includes Beclin 1 and VPS34.The results of these tissue-based analyses provide new insights into sunitinib"s mechanism of action in SFT patients. Sunitinib 175-184 platelet derived growth factor receptor beta Homo sapiens 28-34 27304187-8 2016 Finally, we also found that PDGFRB is one of the components of a complex that includes Beclin 1 and VPS34.The results of these tissue-based analyses provide new insights into sunitinib"s mechanism of action in SFT patients. Sunitinib 175-184 beclin 1 Homo sapiens 87-95 27304187-8 2016 Finally, we also found that PDGFRB is one of the components of a complex that includes Beclin 1 and VPS34.The results of these tissue-based analyses provide new insights into sunitinib"s mechanism of action in SFT patients. Sunitinib 175-184 phosphatidylinositol 3-kinase catalytic subunit type 3 Homo sapiens 100-105 27181063-7 2016 Although the patient had refractory disease after salvage therapy for the second relapse, treatment with the FLT3 inhibitor sunitinib rapidly induced a near complete molecular response, permitting the patient to proceed to a matched-unrelated donor stem cell transplantation. Sunitinib 124-133 fms related receptor tyrosine kinase 3 Homo sapiens 109-113 27109549-1 2016 Sunitinib malate is a small multi-targeted tyrosine kinase inhibitor that inhibits vascular endothelial growth factor receptor (VEGFR), platelet-derived growth factor receptor (PDGFR) and stem cell factor receptor (KIT), which are highly expressed by some high-grade brain tumors. Sunitinib 0-16 kinase insert domain receptor Homo sapiens 83-126 27109549-1 2016 Sunitinib malate is a small multi-targeted tyrosine kinase inhibitor that inhibits vascular endothelial growth factor receptor (VEGFR), platelet-derived growth factor receptor (PDGFR) and stem cell factor receptor (KIT), which are highly expressed by some high-grade brain tumors. Sunitinib 0-16 kinase insert domain receptor Homo sapiens 128-133 27109549-1 2016 Sunitinib malate is a small multi-targeted tyrosine kinase inhibitor that inhibits vascular endothelial growth factor receptor (VEGFR), platelet-derived growth factor receptor (PDGFR) and stem cell factor receptor (KIT), which are highly expressed by some high-grade brain tumors. Sunitinib 0-16 platelet derived growth factor receptor beta Homo sapiens 136-175 27109549-1 2016 Sunitinib malate is a small multi-targeted tyrosine kinase inhibitor that inhibits vascular endothelial growth factor receptor (VEGFR), platelet-derived growth factor receptor (PDGFR) and stem cell factor receptor (KIT), which are highly expressed by some high-grade brain tumors. Sunitinib 0-16 platelet derived growth factor receptor beta Homo sapiens 177-182 27109549-1 2016 Sunitinib malate is a small multi-targeted tyrosine kinase inhibitor that inhibits vascular endothelial growth factor receptor (VEGFR), platelet-derived growth factor receptor (PDGFR) and stem cell factor receptor (KIT), which are highly expressed by some high-grade brain tumors. Sunitinib 0-16 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 215-218 27109549-9 2016 While there was a statistically significant modulation of plasma VEGFR2 with sunitinib exposure, there were no sustained tumor responses. Sunitinib 77-86 kinase insert domain receptor Homo sapiens 65-71 27156227-10 2016 Nine patients (35%) had a best response of stable disease, including two patients who had KIT mutations known to be associated with resistance to imatinib and sunitinib. Sunitinib 159-168 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 90-93 27479949-10 2016 Treatment of EHE with VEGF inhibition, potentially in combination with other antiangiogenic and tumor-inhibiting therapies such as lenalidomide, thalidomide, sorafenib, and sunitinib, may also hold promise. Sunitinib 173-182 vascular endothelial growth factor A Homo sapiens 22-26 30058926-9 2016 Targeted agents, such as everolimus and sunitinib, have improved PFS in GEP-NET. Sunitinib 40-49 granulin precursor Homo sapiens 72-75 27564279-0 2016 Sunitinib treatment in patients with advanced renal cell cancer: the Brazilian National Cancer Institute (INCA) experience. Sunitinib 0-9 caspase recruitment domain family member 17 Homo sapiens 106-110 27237035-1 2016 BACKGROUND: Sunitinib is a potent oral tyrosine kinase inhibitor of VEGFRs, KIT, and PDGFRs. Sunitinib 12-21 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 76-79 27385210-0 2016 Sunitinib enhances the antitumor responses of agonistic CD40-antibody by reducing MDSCs and synergistically improving endothelial activation and T-cell recruitment. Sunitinib 0-9 CD40 molecule Homo sapiens 56-60 27385210-4 2016 Treatment of tumor-bearing mice with anti-CD40 mAb led to increased activation of CD11c+ dendritic cells in the tumor draining lymph node, while sunitinib treatment reduced vessel density and decreased accumulation of CD11b+Gr1+ myeloid derived suppressor cells. Sunitinib 145-154 integrin subunit alpha M Homo sapiens 218-223 27385210-5 2016 The expression of ICAM-1 and VCAM-1 adhesion molecules was up-regulated on tumor endothelial cells only when anti-CD40 mAb treatment was combined with sunitinib. Sunitinib 151-160 intercellular adhesion molecule 1 Homo sapiens 18-24 27385210-5 2016 The expression of ICAM-1 and VCAM-1 adhesion molecules was up-regulated on tumor endothelial cells only when anti-CD40 mAb treatment was combined with sunitinib. Sunitinib 151-160 vascular cell adhesion molecule 1 Homo sapiens 29-35 27385210-7 2016 Our results show that combining CD40-stimulating immunotherapy with sunitinib treatment exerts potent complementary antitumor effects mediated by dendritic cell activation, a reduction in myeloid derived suppressor cells and increased endothelial activation, resulting in enhanced recruitment of cytotoxic T-cells. Sunitinib 68-77 CD40 molecule Homo sapiens 32-36 27175586-2 2016 We previously reported how VEGF and VEGFR polymorphisms might have a predictive role in patients treated with first-line sunitinib. Sunitinib 121-130 vascular endothelial growth factor A Homo sapiens 27-31 26733173-6 2016 EGCG enhanced the antiproliferation and VEGF secretion-reducing effects of sunitinib in the three tested cell lines. Sunitinib 75-84 vascular endothelial growth factor A Homo sapiens 40-44 26733173-8 2016 We further demonstrated that sunitinib exposure induces insulin receptor substrate-1 (IRS-1) upregulation and activation of MAPK signaling. Sunitinib 29-38 insulin receptor substrate 1 Homo sapiens 56-84 26733173-8 2016 We further demonstrated that sunitinib exposure induces insulin receptor substrate-1 (IRS-1) upregulation and activation of MAPK signaling. Sunitinib 29-38 insulin receptor substrate 1 Homo sapiens 86-91 27175586-2 2016 We previously reported how VEGF and VEGFR polymorphisms might have a predictive role in patients treated with first-line sunitinib. Sunitinib 121-130 kinase insert domain receptor Homo sapiens 36-41 27175586-7 2016 CONCLUSIONS: in our analysis patients with opposite polymorphisms of rs833061, rs2010963, rs699947 of VEGF A seems to have a better PFS if treated with either sunitinib or pazopanib. Sunitinib 159-168 vascular endothelial growth factor A Homo sapiens 102-108 27770111-0 2016 Correction: Sunitinib prevents cachexia and prolongs survival of mice bearing renal cancer by restraining STAT3 and MuRF-1 activation in muscle. Sunitinib 12-21 signal transducer and activator of transcription 3 Mus musculus 106-111 27141054-7 2016 Sunitinib sensitivity correlated with vascular endothelial growth factor (VEGF) production. Sunitinib 0-9 vascular endothelial growth factor A Homo sapiens 38-72 27770111-0 2016 Correction: Sunitinib prevents cachexia and prolongs survival of mice bearing renal cancer by restraining STAT3 and MuRF-1 activation in muscle. Sunitinib 12-21 tripartite motif-containing 63 Mus musculus 116-122 26815723-0 2016 Sunitinib activates Axl signaling in renal cell cancer. Sunitinib 0-9 AXL receptor tyrosine kinase Homo sapiens 20-23 26815723-7 2016 Compared to control, incubation with sunitinib at its IC50 of 2 microM resulted in downregulation of 86 phosphopeptides including CDK5, DYRK3, DYRK4, G6PD, PKM and LDH-A, while 94 phosphopeptides including Axl, FAK, EPHA2 and p38alpha were upregulated. Sunitinib 37-46 cyclin dependent kinase 5 Homo sapiens 130-134 26815723-7 2016 Compared to control, incubation with sunitinib at its IC50 of 2 microM resulted in downregulation of 86 phosphopeptides including CDK5, DYRK3, DYRK4, G6PD, PKM and LDH-A, while 94 phosphopeptides including Axl, FAK, EPHA2 and p38alpha were upregulated. Sunitinib 37-46 dual specificity tyrosine phosphorylation regulated kinase 3 Homo sapiens 136-141 26815723-7 2016 Compared to control, incubation with sunitinib at its IC50 of 2 microM resulted in downregulation of 86 phosphopeptides including CDK5, DYRK3, DYRK4, G6PD, PKM and LDH-A, while 94 phosphopeptides including Axl, FAK, EPHA2 and p38alpha were upregulated. Sunitinib 37-46 dual specificity tyrosine phosphorylation regulated kinase 4 Homo sapiens 143-148 26815723-7 2016 Compared to control, incubation with sunitinib at its IC50 of 2 microM resulted in downregulation of 86 phosphopeptides including CDK5, DYRK3, DYRK4, G6PD, PKM and LDH-A, while 94 phosphopeptides including Axl, FAK, EPHA2 and p38alpha were upregulated. Sunitinib 37-46 glucose-6-phosphate dehydrogenase Homo sapiens 150-154 26815723-7 2016 Compared to control, incubation with sunitinib at its IC50 of 2 microM resulted in downregulation of 86 phosphopeptides including CDK5, DYRK3, DYRK4, G6PD, PKM and LDH-A, while 94 phosphopeptides including Axl, FAK, EPHA2 and p38alpha were upregulated. Sunitinib 37-46 pyruvate kinase M1/2 Homo sapiens 156-159 26815723-7 2016 Compared to control, incubation with sunitinib at its IC50 of 2 microM resulted in downregulation of 86 phosphopeptides including CDK5, DYRK3, DYRK4, G6PD, PKM and LDH-A, while 94 phosphopeptides including Axl, FAK, EPHA2 and p38alpha were upregulated. Sunitinib 37-46 lactate dehydrogenase A Homo sapiens 164-169 26815723-7 2016 Compared to control, incubation with sunitinib at its IC50 of 2 microM resulted in downregulation of 86 phosphopeptides including CDK5, DYRK3, DYRK4, G6PD, PKM and LDH-A, while 94 phosphopeptides including Axl, FAK, EPHA2 and p38alpha were upregulated. Sunitinib 37-46 AXL receptor tyrosine kinase Homo sapiens 206-209 26815723-7 2016 Compared to control, incubation with sunitinib at its IC50 of 2 microM resulted in downregulation of 86 phosphopeptides including CDK5, DYRK3, DYRK4, G6PD, PKM and LDH-A, while 94 phosphopeptides including Axl, FAK, EPHA2 and p38alpha were upregulated. Sunitinib 37-46 protein tyrosine kinase 2 Homo sapiens 211-214 26815723-7 2016 Compared to control, incubation with sunitinib at its IC50 of 2 microM resulted in downregulation of 86 phosphopeptides including CDK5, DYRK3, DYRK4, G6PD, PKM and LDH-A, while 94 phosphopeptides including Axl, FAK, EPHA2 and p38alpha were upregulated. Sunitinib 37-46 EPH receptor A2 Homo sapiens 216-221 26815723-7 2016 Compared to control, incubation with sunitinib at its IC50 of 2 microM resulted in downregulation of 86 phosphopeptides including CDK5, DYRK3, DYRK4, G6PD, PKM and LDH-A, while 94 phosphopeptides including Axl, FAK, EPHA2 and p38alpha were upregulated. Sunitinib 37-46 mitogen-activated protein kinase 14 Homo sapiens 226-234 26815723-9 2016 Subsequent proliferation assays revealed that inhibition of Axl with a small molecule inhibitor (R428) sensitized 786-O RCC cells and immortalized endothelial cells to sunitinib up to 3 fold. Sunitinib 168-177 AXL receptor tyrosine kinase Homo sapiens 60-63 27141054-7 2016 Sunitinib sensitivity correlated with vascular endothelial growth factor (VEGF) production. Sunitinib 0-9 vascular endothelial growth factor A Homo sapiens 74-78 26815723-10 2016 In conclusion, incubation with sunitinib of RCC cells causes significant upregulation of multiple phosphopeptides including Axl. Sunitinib 31-40 AXL receptor tyrosine kinase Homo sapiens 124-127 26815723-11 2016 Simultaneous inhibition of Axl improves the antitumor activity of sunitinib. Sunitinib 66-75 AXL receptor tyrosine kinase Homo sapiens 27-30 27141054-8 2016 RCC induced paracrine extracellular signal-regulated kinase (ERK) activation in EC which was inhibited by sunitinib in sensitive but not in resistant tumors. Sunitinib 106-115 mitogen-activated protein kinase 1 Homo sapiens 22-59 27141054-8 2016 RCC induced paracrine extracellular signal-regulated kinase (ERK) activation in EC which was inhibited by sunitinib in sensitive but not in resistant tumors. Sunitinib 106-115 mitogen-activated protein kinase 1 Homo sapiens 61-64 27141054-11 2016 Our data show that RCC activates EC through VEGF-dependent and -independent pathways, that sunitinib sensitivity correlates with VEGF-mediated ERK activation, and that combined inhibition of VEGF/PDGF/FGF receptors is sufficient to inhibit mitogenic signaling in EC but not in fibroblasts. Sunitinib 91-100 vascular endothelial growth factor A Homo sapiens 129-133 27141054-11 2016 Our data show that RCC activates EC through VEGF-dependent and -independent pathways, that sunitinib sensitivity correlates with VEGF-mediated ERK activation, and that combined inhibition of VEGF/PDGF/FGF receptors is sufficient to inhibit mitogenic signaling in EC but not in fibroblasts. Sunitinib 91-100 mitogen-activated protein kinase 1 Homo sapiens 143-146 27141054-11 2016 Our data show that RCC activates EC through VEGF-dependent and -independent pathways, that sunitinib sensitivity correlates with VEGF-mediated ERK activation, and that combined inhibition of VEGF/PDGF/FGF receptors is sufficient to inhibit mitogenic signaling in EC but not in fibroblasts. Sunitinib 91-100 vascular endothelial growth factor A Homo sapiens 129-133 27103123-1 2016 PURPOSE: Sirolimus, an oral mTOR inhibitor, may complement the anti-angiogenic and anti-tumor activity of sunitinib, an oral small molecule inhibitor of multiple receptor tyrosine kinases, by vertical disruption of vascular epithelial growth factor receptor (VEGFR) signaling, by reducing the compensatory production of VEGF in sunitinib-treated patients and also by directly inhibiting tumor cell proliferation. Sunitinib 106-115 mechanistic target of rapamycin kinase Homo sapiens 28-32 27103123-1 2016 PURPOSE: Sirolimus, an oral mTOR inhibitor, may complement the anti-angiogenic and anti-tumor activity of sunitinib, an oral small molecule inhibitor of multiple receptor tyrosine kinases, by vertical disruption of vascular epithelial growth factor receptor (VEGFR) signaling, by reducing the compensatory production of VEGF in sunitinib-treated patients and also by directly inhibiting tumor cell proliferation. Sunitinib 106-115 kinase insert domain receptor Homo sapiens 215-257 27103123-1 2016 PURPOSE: Sirolimus, an oral mTOR inhibitor, may complement the anti-angiogenic and anti-tumor activity of sunitinib, an oral small molecule inhibitor of multiple receptor tyrosine kinases, by vertical disruption of vascular epithelial growth factor receptor (VEGFR) signaling, by reducing the compensatory production of VEGF in sunitinib-treated patients and also by directly inhibiting tumor cell proliferation. Sunitinib 106-115 kinase insert domain receptor Homo sapiens 259-264 27103123-1 2016 PURPOSE: Sirolimus, an oral mTOR inhibitor, may complement the anti-angiogenic and anti-tumor activity of sunitinib, an oral small molecule inhibitor of multiple receptor tyrosine kinases, by vertical disruption of vascular epithelial growth factor receptor (VEGFR) signaling, by reducing the compensatory production of VEGF in sunitinib-treated patients and also by directly inhibiting tumor cell proliferation. Sunitinib 106-115 vascular endothelial growth factor A Homo sapiens 259-263 27149458-6 2016 Gene mutations in the genes encoding EGFR, FGFR2, KDR, and RET were discovered in the patient"s tumor tissue by whole exome sequencing and the patient was treated with a combination of the targeted drugs cetuximab and sunitinib. Sunitinib 218-227 epidermal growth factor receptor Homo sapiens 37-41 27300260-5 2016 The results suggest that 1) active drug concentration (i.e., sunitinib and its metabolite) inhibits the release of sVEGFR2 and that such inhibition is associated with TGI, and 2) daily sVEGFR2 exposure is likely a reliable predictor for the TTP in HCC patients. Sunitinib 61-70 ZFP36 ring finger protein Homo sapiens 241-244 26626617-7 2016 At final analysis, mOS was 16.2 and 14.9 mo with sunitinib and everolimus, respectively (p=0.18). Sunitinib 49-58 Moloney sarcoma oncogene Mus musculus 19-22 26626617-9 2016 In patients without sarcomatoid features in their tumors (n=49), mOS was 31.6 mo with sunitinib and 10.5 mo with everolimus (p=0.075). Sunitinib 86-95 Moloney sarcoma oncogene Mus musculus 65-68 26681729-0 2016 Effect of high salt diet on blood pressure and renal damage during vascular endothelial growth factor inhibition with sunitinib. Sunitinib 118-127 vascular endothelial growth factor A Rattus norvegicus 67-101 26681729-1 2016 BACKGROUND: Antiangiogenic treatment with the multitargeted vascular endothelial growth factor (VEGF) receptor inhibitor sunitinib associates with a blood pressure (BP) rise and glomerular renal injury. Sunitinib 121-130 vascular endothelial growth factor A Rattus norvegicus 60-94 26681729-1 2016 BACKGROUND: Antiangiogenic treatment with the multitargeted vascular endothelial growth factor (VEGF) receptor inhibitor sunitinib associates with a blood pressure (BP) rise and glomerular renal injury. Sunitinib 121-130 vascular endothelial growth factor A Rattus norvegicus 96-100 27117758-4 2016 lncARSR promoted sunitinib resistance via competitively binding miR-34/miR-449 to facilitate AXL and c-MET expression in RCC cells. Sunitinib 17-26 microRNA 34a Homo sapiens 64-70 27117758-4 2016 lncARSR promoted sunitinib resistance via competitively binding miR-34/miR-449 to facilitate AXL and c-MET expression in RCC cells. Sunitinib 17-26 AXL receptor tyrosine kinase Homo sapiens 93-96 27117758-4 2016 lncARSR promoted sunitinib resistance via competitively binding miR-34/miR-449 to facilitate AXL and c-MET expression in RCC cells. Sunitinib 17-26 MET proto-oncogene, receptor tyrosine kinase Homo sapiens 101-106 27117758-6 2016 Treatment of sunitinib-resistant RCC with locked nucleic acids targeting lncARSR or an AXL/c-MET inhibitor restored sunitinib response. Sunitinib 13-22 AXL receptor tyrosine kinase Homo sapiens 87-90 27117758-6 2016 Treatment of sunitinib-resistant RCC with locked nucleic acids targeting lncARSR or an AXL/c-MET inhibitor restored sunitinib response. Sunitinib 116-125 AXL receptor tyrosine kinase Homo sapiens 87-90 27117758-6 2016 Treatment of sunitinib-resistant RCC with locked nucleic acids targeting lncARSR or an AXL/c-MET inhibitor restored sunitinib response. Sunitinib 116-125 MET proto-oncogene, receptor tyrosine kinase Homo sapiens 91-96 27149458-6 2016 Gene mutations in the genes encoding EGFR, FGFR2, KDR, and RET were discovered in the patient"s tumor tissue by whole exome sequencing and the patient was treated with a combination of the targeted drugs cetuximab and sunitinib. Sunitinib 218-227 fibroblast growth factor receptor 2 Homo sapiens 43-48 27149458-6 2016 Gene mutations in the genes encoding EGFR, FGFR2, KDR, and RET were discovered in the patient"s tumor tissue by whole exome sequencing and the patient was treated with a combination of the targeted drugs cetuximab and sunitinib. Sunitinib 218-227 kinase insert domain receptor Homo sapiens 50-53 27149458-6 2016 Gene mutations in the genes encoding EGFR, FGFR2, KDR, and RET were discovered in the patient"s tumor tissue by whole exome sequencing and the patient was treated with a combination of the targeted drugs cetuximab and sunitinib. Sunitinib 218-227 ret proto-oncogene Homo sapiens 59-62 26959741-7 2016 PKM2 was also found to determine the sensitivity of targeted drugs, such as sorafenib, brivanib, and sunitinib, by SHP-1 activation. Sunitinib 101-110 pyruvate kinase M1/2 Homo sapiens 0-4 26434595-4 2016 describe the mechanisms linking Axl and Met activation to acquired resistance to sunitinib in renal cell carcinoma (RCC), providing a pre-clinical rationale for the development of Axl and Met inhibitors including cabozantinib in anti-angiogenic resistant RCC. Sunitinib 81-90 AXL receptor tyrosine kinase Homo sapiens 180-183 27077705-2 2016 Clinically used c-KIT kinase inhibitors, i.e., Imatinib and Sunitinib, bear other important targets such as ABL or FLT3 kinases. Sunitinib 60-69 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 16-21 27100185-0 2016 Association of Angiopoietin-2 and Ki-67 Expression with Vascular Density and Sunitinib Response in Metastatic Renal Cell Carcinoma. Sunitinib 77-86 angiopoietin 2 Homo sapiens 15-29 27100185-5 2016 Ang2 protein expression was restrained to RCC tumour vessels, and correlated with tumour vascularization and response to sunitinib. Sunitinib 121-130 angiopoietin 2 Homo sapiens 0-4 27100185-8 2016 In summary, in this study to investigate endothelial Ang2 in mRCC patients treated with first-line sunitinib, high cancer Ang2 expression was associated with the CBR, but not PFS or OS, whereas low Ki-67 expression was significantly associated with long PFS and OS. Sunitinib 99-108 angiopoietin 2 Homo sapiens 53-57 26364599-0 2016 Targeting MET and AXL overcomes resistance to sunitinib therapy in renal cell carcinoma. Sunitinib 46-55 AXL receptor tyrosine kinase Homo sapiens 18-21 26364599-3 2016 We hypothesized that sunitinib-induced upregulation of the prometastatic MET and AXL receptors is associated with resistance to sunitinib and with more aggressive tumor behavior. Sunitinib 21-30 AXL receptor tyrosine kinase Homo sapiens 81-84 26364599-3 2016 We hypothesized that sunitinib-induced upregulation of the prometastatic MET and AXL receptors is associated with resistance to sunitinib and with more aggressive tumor behavior. Sunitinib 128-137 AXL receptor tyrosine kinase Homo sapiens 81-84 26364599-10 2016 Chronic sunitinib treatment of RCC cell lines activated both AXL and MET, induced EMT-associated gene expression changes, including upregulation of Snail and beta-catenin, and increased cell migration and invasion. Sunitinib 8-17 AXL receptor tyrosine kinase Homo sapiens 61-64 26364599-10 2016 Chronic sunitinib treatment of RCC cell lines activated both AXL and MET, induced EMT-associated gene expression changes, including upregulation of Snail and beta-catenin, and increased cell migration and invasion. Sunitinib 8-17 catenin beta 1 Homo sapiens 158-170 26364599-12 2016 The suppression of AXL or MET expression and the inhibition of AXL and MET activation using cabozantinib both impaired chronic sunitinib treatment-induced prometastatic behavior in cell culture and rescued acquired resistance to sunitinib in xenograft models. Sunitinib 127-136 AXL receptor tyrosine kinase Homo sapiens 63-66 26364599-12 2016 The suppression of AXL or MET expression and the inhibition of AXL and MET activation using cabozantinib both impaired chronic sunitinib treatment-induced prometastatic behavior in cell culture and rescued acquired resistance to sunitinib in xenograft models. Sunitinib 229-238 AXL receptor tyrosine kinase Homo sapiens 63-66 26364599-13 2016 In summary, chronic sunitinib treatment induces the activation of AXL and MET signaling and promotes prometastatic behavior and angiogenesis. Sunitinib 20-29 AXL receptor tyrosine kinase Homo sapiens 66-69 26364599-14 2016 The inhibition of AXL and MET activity may overcome resistance induced by prolonged sunitinib therapy in metastatic RCC. Sunitinib 84-93 AXL receptor tyrosine kinase Homo sapiens 18-21 26959741-7 2016 PKM2 was also found to determine the sensitivity of targeted drugs, such as sorafenib, brivanib, and sunitinib, by SHP-1 activation. Sunitinib 101-110 protein tyrosine phosphatase non-receptor type 6 Homo sapiens 115-120 26934000-5 2016 The combination of the ERBB1/2/4 inhibitor afatinib with the SRC family inhibitor dasatinib killed afatinib resistant H1975 cells in a greater than additive fashion; other drugs used in combination with dasatinib such as sunitinib, crizotinib and amufatinib were less effective. Sunitinib 221-230 epidermal growth factor receptor Homo sapiens 23-32 26447463-0 2016 The early effect of sunitinib on insulin clearance in patients with metastatic renal cell carcinoma. Sunitinib 20-29 insulin Homo sapiens 33-40 26896780-3 2016 In this study, protein-ligand interactions between FLT3 and kinase inhibitors (CEP701, PKC412, sunitinib, imatinib and dasatinib) were obtained through homology modelling and molecular docking. Sunitinib 95-104 FMS-like tyrosine kinase 3 Mus musculus 51-55 26896780-6 2016 CEP701, PKC412 and sunitinib interacted with the ATP-binding pocket of FLT3, forming H-bonds with Cys694 and Glu692. Sunitinib 19-28 FMS-like tyrosine kinase 3 Mus musculus 71-75 26896780-9 2016 The potency of sunitinib and to a lesser extent CEP701 in inhibition of FLT3 autophosphorylation was lower than the cell proliferation inhibition, indicating that inhibition of FLT3 downstream proteins may contribute to the cellular effects. Sunitinib 15-24 FMS-like tyrosine kinase 3 Mus musculus 72-76 26896780-9 2016 The potency of sunitinib and to a lesser extent CEP701 in inhibition of FLT3 autophosphorylation was lower than the cell proliferation inhibition, indicating that inhibition of FLT3 downstream proteins may contribute to the cellular effects. Sunitinib 15-24 FMS-like tyrosine kinase 3 Mus musculus 177-181 26912111-1 2016 Sunitinib (Sutent ) is a receptor tyrosine kinase (RTK) and angiogenesis inhibitor approved for the treatment of renal cell carcinomas, gastrointestinal stromal tumours and pancreatic neuroendocrine tumours. Sunitinib 0-9 erb-b2 receptor tyrosine kinase 4 Rattus norvegicus 25-49 26912111-1 2016 Sunitinib (Sutent ) is a receptor tyrosine kinase (RTK) and angiogenesis inhibitor approved for the treatment of renal cell carcinomas, gastrointestinal stromal tumours and pancreatic neuroendocrine tumours. Sunitinib 0-9 erb-b2 receptor tyrosine kinase 4 Rattus norvegicus 51-54 26912111-1 2016 Sunitinib (Sutent ) is a receptor tyrosine kinase (RTK) and angiogenesis inhibitor approved for the treatment of renal cell carcinomas, gastrointestinal stromal tumours and pancreatic neuroendocrine tumours. Sunitinib 11-17 erb-b2 receptor tyrosine kinase 4 Rattus norvegicus 25-49 26912111-1 2016 Sunitinib (Sutent ) is a receptor tyrosine kinase (RTK) and angiogenesis inhibitor approved for the treatment of renal cell carcinomas, gastrointestinal stromal tumours and pancreatic neuroendocrine tumours. Sunitinib 11-17 erb-b2 receptor tyrosine kinase 4 Rattus norvegicus 51-54 26447463-2 2016 To explore the mechanism of this adverse effect we performed a prospective study to investigate the effect of sunitinib on insulin concentration, insulin clearance and insulin sensitivity. Sunitinib 110-119 insulin Homo sapiens 123-130 26447463-6 2016 Steady-state insulin concentrations during the clamp increased after 1 week of sunitinib (from 128.9 +- 9.0 mU l-1 to 170.8 +- 12.8 mU l-1, P < 0.05; 95% CI on difference - 64.3, -19.6). Sunitinib 79-88 insulin Homo sapiens 13-20 26447463-9 2016 CONCLUSION: Sunitinib affects insulin clearance which could possibly lead to overexposure to insulin in patients using insulin or insulin-secretion stimulating agents. Sunitinib 12-21 insulin Homo sapiens 30-37 26447463-9 2016 CONCLUSION: Sunitinib affects insulin clearance which could possibly lead to overexposure to insulin in patients using insulin or insulin-secretion stimulating agents. Sunitinib 12-21 insulin Homo sapiens 93-100 26447463-9 2016 CONCLUSION: Sunitinib affects insulin clearance which could possibly lead to overexposure to insulin in patients using insulin or insulin-secretion stimulating agents. Sunitinib 12-21 insulin Homo sapiens 93-100 26447463-9 2016 CONCLUSION: Sunitinib affects insulin clearance which could possibly lead to overexposure to insulin in patients using insulin or insulin-secretion stimulating agents. Sunitinib 12-21 insulin Homo sapiens 93-100 27031198-1 2016 In metastatic ccRCC , the treatment options in 1st line treatment are still the tyrosinkinase inhibitors (TKI) pazopanib and sunitinib, for patients with low or intermediate risk additionally IFNalpha/bevacizumab and for high risk patients the mTOR inhibitor temsirolimus. Sunitinib 125-134 mechanistic target of rapamycin kinase Homo sapiens 244-248 27453754-4 2016 Sunitinib represents an inhibitor of VEGFR 1-3, c-kit, FLT-3 and PDGFR. Sunitinib 0-9 fms related receptor tyrosine kinase 1 Homo sapiens 37-46 27453754-4 2016 Sunitinib represents an inhibitor of VEGFR 1-3, c-kit, FLT-3 and PDGFR. Sunitinib 0-9 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 48-53 27453754-4 2016 Sunitinib represents an inhibitor of VEGFR 1-3, c-kit, FLT-3 and PDGFR. Sunitinib 0-9 fms related receptor tyrosine kinase 3 Homo sapiens 55-60 27453754-4 2016 Sunitinib represents an inhibitor of VEGFR 1-3, c-kit, FLT-3 and PDGFR. Sunitinib 0-9 platelet derived growth factor receptor beta Homo sapiens 65-70 27073655-4 2016 A patient with a c-kit mutation-positive thymic carcinoma received imatinib followed by sunitinib consecutively, which are both c-Kit inhibitors. Sunitinib 88-97 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 17-22 27073655-4 2016 A patient with a c-kit mutation-positive thymic carcinoma received imatinib followed by sunitinib consecutively, which are both c-Kit inhibitors. Sunitinib 88-97 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 128-133 27034725-2 2016 Improvements in response rates and survival, with more manageable side effects compared with interleukin 2/interferon immunotherapy, have been reported with the use of targeted therapy agents, including vascular endothelial growth factor (VEGF) receptor tyrosine kinase inhibitors (sunitinib, sorafenib, pazopanib, axitinib), mammalian target of rapamycin (mTOR) inhibitors (everolimus and temsirolimus) and VEGF receptor antibodies (bevacizumab). Sunitinib 282-291 vascular endothelial growth factor A Homo sapiens 203-237 27035144-4 2016 Sunitinib exposure significantly reduced (i) corpora lutea number per ovary (1.1 +- 0.38 in sunitinib group versus 4 +- 0.79 in control group, p<0.01) and (ii) serum Anti Mullerian hormone (AMH) levels in sunitinib treated mice (12.01 +- 1.16) compared to control mice (14.33 +- 0.87 ng/ml, p< 0.05). Sunitinib 0-9 anti-Mullerian hormone Mus musculus 193-196 27053605-8 2016 CONCLUSION: Sunitinib inhibits the expressions of CD80, PD-L1 and B7-H4 on DCs induced by LPS. Sunitinib 12-21 CD80 molecule Homo sapiens 50-54 27053605-8 2016 CONCLUSION: Sunitinib inhibits the expressions of CD80, PD-L1 and B7-H4 on DCs induced by LPS. Sunitinib 12-21 CD274 molecule Homo sapiens 56-61 27053605-7 2016 Compared with the LPS-treated group, the expressions of CD80, PD-L1 and B7-H4 on DCs significantly decreased in the sunitinib-LPS group; the expressions of PD-L1, PD-L2, CD80, CD86, B7-H4 and HVEM were lower in the DMSO group. Sunitinib 116-125 CD80 molecule Homo sapiens 56-60 27053605-7 2016 Compared with the LPS-treated group, the expressions of CD80, PD-L1 and B7-H4 on DCs significantly decreased in the sunitinib-LPS group; the expressions of PD-L1, PD-L2, CD80, CD86, B7-H4 and HVEM were lower in the DMSO group. Sunitinib 116-125 CD274 molecule Homo sapiens 62-67 27053605-7 2016 Compared with the LPS-treated group, the expressions of CD80, PD-L1 and B7-H4 on DCs significantly decreased in the sunitinib-LPS group; the expressions of PD-L1, PD-L2, CD80, CD86, B7-H4 and HVEM were lower in the DMSO group. Sunitinib 116-125 CD274 molecule Homo sapiens 156-161 27053605-7 2016 Compared with the LPS-treated group, the expressions of CD80, PD-L1 and B7-H4 on DCs significantly decreased in the sunitinib-LPS group; the expressions of PD-L1, PD-L2, CD80, CD86, B7-H4 and HVEM were lower in the DMSO group. Sunitinib 116-125 programmed cell death 1 ligand 2 Homo sapiens 163-168 27053605-7 2016 Compared with the LPS-treated group, the expressions of CD80, PD-L1 and B7-H4 on DCs significantly decreased in the sunitinib-LPS group; the expressions of PD-L1, PD-L2, CD80, CD86, B7-H4 and HVEM were lower in the DMSO group. Sunitinib 116-125 CD80 molecule Homo sapiens 170-174 27053605-7 2016 Compared with the LPS-treated group, the expressions of CD80, PD-L1 and B7-H4 on DCs significantly decreased in the sunitinib-LPS group; the expressions of PD-L1, PD-L2, CD80, CD86, B7-H4 and HVEM were lower in the DMSO group. Sunitinib 116-125 CD86 molecule Homo sapiens 176-180 27053605-7 2016 Compared with the LPS-treated group, the expressions of CD80, PD-L1 and B7-H4 on DCs significantly decreased in the sunitinib-LPS group; the expressions of PD-L1, PD-L2, CD80, CD86, B7-H4 and HVEM were lower in the DMSO group. Sunitinib 116-125 TNF receptor superfamily member 14 Homo sapiens 192-196 27034725-2 2016 Improvements in response rates and survival, with more manageable side effects compared with interleukin 2/interferon immunotherapy, have been reported with the use of targeted therapy agents, including vascular endothelial growth factor (VEGF) receptor tyrosine kinase inhibitors (sunitinib, sorafenib, pazopanib, axitinib), mammalian target of rapamycin (mTOR) inhibitors (everolimus and temsirolimus) and VEGF receptor antibodies (bevacizumab). Sunitinib 282-291 vascular endothelial growth factor A Homo sapiens 239-243 26786874-5 2016 We show that frequencies of monocytic CD14(+)HLA-DR(-/low) MDSCs derived from mature monocytes were significantly and dose-dependently reduced in the presence of dasatinib, nilotinib and sorafenib, whereas sunitinib had no effect. Sunitinib 206-215 CD14 molecule Homo sapiens 38-42 26923603-1 2016 Autocrine VEGF signaling is critical for sustaining prostate and other cancer stem cells (CSCs), and it is a potential therapeutic target, but we observed that CSCs isolated from prostate tumors are resistant to anti-VEGF (bevacizumab) and anti-VEGFR (sunitinib) therapy. Sunitinib 252-261 vascular endothelial growth factor A Homo sapiens 10-14 26832420-2 2016 Oral tyrosine kinase inhibitors (TKI) targeting the VEGF receptor, including sunitinib, sorafenib, axitinib, regorafenib, pazopanib, and vandetanib reduce tumor growth and metastasis. Sunitinib 77-86 vascular endothelial growth factor A Homo sapiens 52-56 26914831-0 2016 Association Study of a Functional Variant on ABCG2 Gene with Sunitinib-Induced Severe Adverse Drug Reaction. Sunitinib 61-70 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 45-50 26359568-0 2016 Combined Treatment With 131I-MIBG and Sunitinib Induces Remission in a Patient With Metastatic Paraganglioma Due to Hereditary Paraganglioma-Pheochromocytoma Syndrome From an SDHB Mutation. Sunitinib 38-47 succinate dehydrogenase complex iron sulfur subunit B Homo sapiens 175-179 26914831-3 2016 This study is to investigate the association of a functional germline variant on ABCG2 that affects the pharmacokinetics of sunitinib with sunitinib-induced toxicity of RCC patients in the Japanese population. Sunitinib 124-133 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 81-86 26914831-3 2016 This study is to investigate the association of a functional germline variant on ABCG2 that affects the pharmacokinetics of sunitinib with sunitinib-induced toxicity of RCC patients in the Japanese population. Sunitinib 139-148 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 81-86 26914831-8 2016 In the univariate analysis, both age (P = 7.77x10(-3), odds ratio (OR) = 1.04, 95%CI = 1.01-1.07) and ABCG2 421C>A (P = 1.87x10(-2), OR = 1.71, 95%CI = 1.09-2.68) showed association with sunitinib-induced severe thrombocytopenia. Sunitinib 190-199 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 102-107 26914831-12 2016 ABCG2 421C>A could explain part of the inter-individual variability of sunitinib-induced severe thrombocytopenia. Sunitinib 74-83 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 0-5 27538132-0 2016 [Sunitinib induces autophagy via suppressing Akt/mTOR pathway in renal cell carcinoma]. Sunitinib 1-10 AKT serine/threonine kinase 1 Homo sapiens 45-48 27538132-0 2016 [Sunitinib induces autophagy via suppressing Akt/mTOR pathway in renal cell carcinoma]. Sunitinib 1-10 mechanistic target of rapamycin kinase Homo sapiens 49-53 27538132-10 2016 And the nuclear-to-cytosol translocation as well as aggregation of LC3-II was presented after sunitinib treatment by the fluorescence microscope, which was the proof of the enhanced autophagy. Sunitinib 94-103 microtubule associated protein 1 light chain 3 alpha Homo sapiens 67-70 27538132-11 2016 According to the immunoblotting, sunitinib was able to increase the accumulation of LC3-II . Sunitinib 33-42 microtubule associated protein 1 light chain 3 alpha Homo sapiens 84-87 27538132-12 2016 At the same time, the result of sunitinib combined with chloroquine, a drug which blocked the fusion of autophagosomes and lysosomes, demonstrated that the increasing amount of LC3-II was due to the enhanced autophagy flux by sunitinib treatment in ACHN cells. Sunitinib 32-41 microtubule associated protein 1 light chain 3 alpha Homo sapiens 177-180 27538132-12 2016 At the same time, the result of sunitinib combined with chloroquine, a drug which blocked the fusion of autophagosomes and lysosomes, demonstrated that the increasing amount of LC3-II was due to the enhanced autophagy flux by sunitinib treatment in ACHN cells. Sunitinib 226-235 microtubule associated protein 1 light chain 3 alpha Homo sapiens 177-180 27538132-14 2016 The rapamycin (mTOR inhibitor) or knocking down Akt subunits could change the sunitinib-induced LC3 -II accumulation, whereas overexpression of Akt subunits decreased the autophagic flux, indicating that Akt/mTOR was the target of sunitinib in autophagy. Sunitinib 78-87 mechanistic target of rapamycin kinase Homo sapiens 15-19 27538132-14 2016 The rapamycin (mTOR inhibitor) or knocking down Akt subunits could change the sunitinib-induced LC3 -II accumulation, whereas overexpression of Akt subunits decreased the autophagic flux, indicating that Akt/mTOR was the target of sunitinib in autophagy. Sunitinib 78-87 AKT serine/threonine kinase 1 Homo sapiens 48-51 27538132-14 2016 The rapamycin (mTOR inhibitor) or knocking down Akt subunits could change the sunitinib-induced LC3 -II accumulation, whereas overexpression of Akt subunits decreased the autophagic flux, indicating that Akt/mTOR was the target of sunitinib in autophagy. Sunitinib 78-87 microtubule associated protein 1 light chain 3 alpha Homo sapiens 96-99 27538132-14 2016 The rapamycin (mTOR inhibitor) or knocking down Akt subunits could change the sunitinib-induced LC3 -II accumulation, whereas overexpression of Akt subunits decreased the autophagic flux, indicating that Akt/mTOR was the target of sunitinib in autophagy. Sunitinib 78-87 mechanistic target of rapamycin kinase Homo sapiens 208-212 27538132-14 2016 The rapamycin (mTOR inhibitor) or knocking down Akt subunits could change the sunitinib-induced LC3 -II accumulation, whereas overexpression of Akt subunits decreased the autophagic flux, indicating that Akt/mTOR was the target of sunitinib in autophagy. Sunitinib 231-240 mechanistic target of rapamycin kinase Homo sapiens 208-212 27538132-15 2016 CONCLUSION: Sunitinib induced autophagy via suppressing Akt/mTOR pathway, and the autophagy was involved in apopotosis. Sunitinib 12-21 AKT serine/threonine kinase 1 Homo sapiens 56-59 27538132-15 2016 CONCLUSION: Sunitinib induced autophagy via suppressing Akt/mTOR pathway, and the autophagy was involved in apopotosis. Sunitinib 12-21 mechanistic target of rapamycin kinase Homo sapiens 60-64 27538133-11 2016 CONCLUSION: MEK/ERK signaling pathways may play an important role in the metastasis and the resistance of sunitinib in RCC patients with bone metastasis. Sunitinib 106-115 mitogen-activated protein kinase kinase 7 Homo sapiens 12-15 27538133-11 2016 CONCLUSION: MEK/ERK signaling pathways may play an important role in the metastasis and the resistance of sunitinib in RCC patients with bone metastasis. Sunitinib 106-115 mitogen-activated protein kinase 1 Homo sapiens 16-19 26831065-3 2016 Aberrant platelet-derived growth factor receptor beta (PDGFRbeta) signaling in cancer has motivated the development of several antagonists currently in clinical use, including imatinib, sunitinib, and sorafenib. Sunitinib 186-195 platelet derived growth factor receptor beta Homo sapiens 9-53 26831065-3 2016 Aberrant platelet-derived growth factor receptor beta (PDGFRbeta) signaling in cancer has motivated the development of several antagonists currently in clinical use, including imatinib, sunitinib, and sorafenib. Sunitinib 186-195 platelet derived growth factor receptor beta Homo sapiens 55-64 26893877-5 2016 Sunitinib is suggested to have preventive effects on the pathogenesis of liver fibrosis and cirrhosis in vitro, via an anti-vascular endothelial growth factor and anti-platelet-derived growth factor mechanism. Sunitinib 0-9 vascular endothelial growth factor A Homo sapiens 124-158 26540173-6 2016 Sunitinib was the most common VEGF-targeted therapy (77%), and it was followed by sorafenib (18.4%). Sunitinib 0-9 vascular endothelial growth factor A Homo sapiens 30-34 26375012-8 2016 RESULTS: Sunitinib suppressed multiple in vitro gain-in-functions of Nf1(+/-) mast cells and fibroblasts and attenuated Erk1/2 phosphorylation. Sunitinib 9-18 neurofibromin 1 Mus musculus 69-72 26375012-8 2016 RESULTS: Sunitinib suppressed multiple in vitro gain-in-functions of Nf1(+/-) mast cells and fibroblasts and attenuated Erk1/2 phosphorylation. Sunitinib 9-18 mitogen-activated protein kinase 3 Mus musculus 120-126 26375012-9 2016 Sunitinib treated Krox20;Nf1(flox/-) mice exhibited significant reductions in pNF size, tumor number, and FDG uptake compared to control mice. Sunitinib 0-9 early growth response 2 Mus musculus 18-24 25778465-0 2016 Effect of the CYP3A5 and ABCB1 genotype on exposure, clinical response and manifestation of toxicities from sunitinib in Asian patients. Sunitinib 108-117 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 14-20 26375012-3 2016 Sunitinib malate is a potent, highly selective RTK inhibitor with activity against c-Kit, PDGFR, and VEGFR, which have also been implicated in the pathogenesis of these lesions. Sunitinib 0-16 KIT proto-oncogene receptor tyrosine kinase Mus musculus 83-88 26375012-3 2016 Sunitinib malate is a potent, highly selective RTK inhibitor with activity against c-Kit, PDGFR, and VEGFR, which have also been implicated in the pathogenesis of these lesions. Sunitinib 0-16 platelet derived growth factor receptor, beta polypeptide Mus musculus 90-95 26375012-9 2016 Sunitinib treated Krox20;Nf1(flox/-) mice exhibited significant reductions in pNF size, tumor number, and FDG uptake compared to control mice. Sunitinib 0-9 neurofibromin 1 Mus musculus 25-28 26375012-5 2016 EXPERIMENTAL DESIGN: Proliferation, beta-hexosaminidase release (degranulation), and Erk1/2 phosphorylation were assessed in sunitinib treated Nf1(+/-) mast cells and fibroblasts, respectively. Sunitinib 125-134 neurofibromin 1 Mus musculus 143-146 25778465-0 2016 Effect of the CYP3A5 and ABCB1 genotype on exposure, clinical response and manifestation of toxicities from sunitinib in Asian patients. Sunitinib 108-117 ATP binding cassette subfamily B member 1 Homo sapiens 25-30 25778465-1 2016 The objective of this study was to determine the effect of the CYP3A5 and ATP binding cassette subfamily B member 1 (ABCB1) single-nucleotide polymorphisms on the disposition of sunitinib and SU12662, on clinical response, and on the manifestation of toxicities in Asian metastatic renal cell carcinoma patients. Sunitinib 178-187 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 63-69 25778465-1 2016 The objective of this study was to determine the effect of the CYP3A5 and ATP binding cassette subfamily B member 1 (ABCB1) single-nucleotide polymorphisms on the disposition of sunitinib and SU12662, on clinical response, and on the manifestation of toxicities in Asian metastatic renal cell carcinoma patients. Sunitinib 178-187 ATP binding cassette subfamily B member 1 Homo sapiens 74-115 26375012-11 2016 CONCLUSIONS: Collectively, these results demonstrate the efficacy of sunitinib in reducing tumor burden in Krox20;Nf1(flox/-) mice. Sunitinib 69-78 early growth response 2 Mus musculus 107-113 25778465-1 2016 The objective of this study was to determine the effect of the CYP3A5 and ATP binding cassette subfamily B member 1 (ABCB1) single-nucleotide polymorphisms on the disposition of sunitinib and SU12662, on clinical response, and on the manifestation of toxicities in Asian metastatic renal cell carcinoma patients. Sunitinib 178-187 ATP binding cassette subfamily B member 1 Homo sapiens 117-122 25778465-5 2016 The CC genotype for ABCB1 was associated with a higher sunitinib exposure (76.81 vs 56.55 ng ml(-1), P=0.03), higher risk of all-grade rash (RR 3.00, 95% CI 1.17-7.67) and mucositis (RR 1.60, 95% CI 1.10-2.34) and disease progression than compared with the CT/TT genotype. Sunitinib 55-64 ATP binding cassette subfamily B member 1 Homo sapiens 20-25 26375012-11 2016 CONCLUSIONS: Collectively, these results demonstrate the efficacy of sunitinib in reducing tumor burden in Krox20;Nf1(flox/-) mice. Sunitinib 69-78 neurofibromin 1 Mus musculus 114-117 26312563-9 2016 S100P knockdown also promoted CCA cell apoptosis by up-regulating expression of apoptosis related factors, DR5, TRADD, caspase 3 and BAX, and increased the sensitivity of CCA cells to the chemotherapeutic agents sunitinib and apigenin. Sunitinib 212-221 S100 calcium binding protein P Homo sapiens 0-5 26772734-0 2016 Correlation of KIT and PDGFRA mutational status with clinical benefit in patients with gastrointestinal stromal tumor treated with sunitinib in a worldwide treatment-use trial. Sunitinib 131-140 platelet derived growth factor receptor alpha Homo sapiens 23-29 26772734-1 2016 BACKGROUND: Several small studies indicated that the genotype of KIT or platelet-derived growth factor receptor-alpha (PDGFRA) contributes in part to the level of clinical effectiveness of sunitinib in gastrointestinal stromal tumor (GIST) patients. Sunitinib 189-198 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 65-68 26772734-1 2016 BACKGROUND: Several small studies indicated that the genotype of KIT or platelet-derived growth factor receptor-alpha (PDGFRA) contributes in part to the level of clinical effectiveness of sunitinib in gastrointestinal stromal tumor (GIST) patients. Sunitinib 189-198 platelet derived growth factor receptor alpha Homo sapiens 72-117 26772734-1 2016 BACKGROUND: Several small studies indicated that the genotype of KIT or platelet-derived growth factor receptor-alpha (PDGFRA) contributes in part to the level of clinical effectiveness of sunitinib in gastrointestinal stromal tumor (GIST) patients. Sunitinib 189-198 platelet derived growth factor receptor alpha Homo sapiens 119-125 26772734-10 2016 The data available were limited to investigate the effects of additional KIT or PDGFRA mutations on the efficacy of sunitinib treatment. Sunitinib 116-125 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 73-76 26772734-10 2016 The data available were limited to investigate the effects of additional KIT or PDGFRA mutations on the efficacy of sunitinib treatment. Sunitinib 116-125 platelet derived growth factor receptor alpha Homo sapiens 80-86 26239999-3 2016 Here, we report that a remarkably synergistic antitumor responses elicited by the combined treatment of Sunitinib and an agonistic antibody against glucocorticoid-induced TNFR related protein (GITR) in a model of metastatic renal cell carcinoma. Sunitinib 104-113 tumor necrosis factor receptor superfamily, member 18 Mus musculus 148-191 25864572-7 2016 RESULTS: VEGFR-2 mRNA expression levels (DeltaCt, median, min-max) were reduced in the everolimus 1.0 (0.25-1.81) and sunitinib 1.06 (0.24-2.68) treated groups compared with the control 4.74 (1.02-14.74) and DMSO groups 7.41 (0.72-13.10). Sunitinib 118-127 kinase insert domain receptor Rattus norvegicus 9-16 26239999-3 2016 Here, we report that a remarkably synergistic antitumor responses elicited by the combined treatment of Sunitinib and an agonistic antibody against glucocorticoid-induced TNFR related protein (GITR) in a model of metastatic renal cell carcinoma. Sunitinib 104-113 tumor necrosis factor receptor superfamily, member 18 Mus musculus 193-197 26239999-4 2016 Sunitinib significantly increased the infiltration, activation, and proliferation and/or cytotoxicity of CD8(+) T cells and NK cells in liver metastatic foci when combined with the anti (alpha)-GITR agonist, which was associated with treatment-induced prominent upregulation of Th1-biased immune genes in the livers from mice receiving combined therapy versus single treatment. Sunitinib 0-9 tumor necrosis factor receptor superfamily, member 18 Mus musculus 194-198 26239999-5 2016 Sunitinib/alpha-GITR treatment also markedly promoted the maturation, activation and cytokine production of liver-resident macrophages and DCs compared with that achieved by alpha-GITR or Sunitinib treatment alone in mice. Sunitinib 0-9 tumor necrosis factor receptor superfamily, member 18 Mus musculus 180-184 26569132-10 2016 Moreover, treatment with bFGF enhanced cell growth and the PDGF-BB-induced migratory activity of cultured pericytes, which were significantly suppressed by SU5402 or Sunitinib, an inhibitor of PDGFR. Sunitinib 166-175 fibroblast growth factor 2 Homo sapiens 25-29 26569132-10 2016 Moreover, treatment with bFGF enhanced cell growth and the PDGF-BB-induced migratory activity of cultured pericytes, which were significantly suppressed by SU5402 or Sunitinib, an inhibitor of PDGFR. Sunitinib 166-175 platelet derived growth factor receptor beta Homo sapiens 193-198 26439451-1 2016 BACKGROUND: Sorafenib and sunitinib are oral vascular endothelial growth factor receptor (VEGFR) tyrosine kinase inhibitors (TKIs) approved in 2005 and 2006, respectively, for the treatment of patients with renal cell carcinoma (RCC). Sunitinib 26-35 kinase insert domain receptor Homo sapiens 45-88 26439451-1 2016 BACKGROUND: Sorafenib and sunitinib are oral vascular endothelial growth factor receptor (VEGFR) tyrosine kinase inhibitors (TKIs) approved in 2005 and 2006, respectively, for the treatment of patients with renal cell carcinoma (RCC). Sunitinib 26-35 kinase insert domain receptor Homo sapiens 90-95 25701259-0 2016 FGF2 Prevents Sunitinib-Induced Cardiotoxicity in Zebrafish and Cardiomyoblast H9c2 Cells. Sunitinib 14-23 fibroblast growth factor 2 Danio rerio 0-4 25701259-4 2016 Here, we reported that the injection of fibroblast growth factor 2 (FGF2) mRNA into one- to two-cell stage embryos protected against sunitinib-induced cardiotoxicity in zebrafish. Sunitinib 133-142 fibroblast growth factor 2 Danio rerio 40-66 25701259-4 2016 Here, we reported that the injection of fibroblast growth factor 2 (FGF2) mRNA into one- to two-cell stage embryos protected against sunitinib-induced cardiotoxicity in zebrafish. Sunitinib 133-142 fibroblast growth factor 2 Danio rerio 68-72 25701259-5 2016 In addition, FGF2 significantly prevented sunitinib-induced cardiotoxicity in cardiomyoblast H9c2 cells, possibly via activating the PLC-gamma/c-Raf/CREB pathway. Sunitinib 42-51 fibroblast growth factor 2 Rattus norvegicus 13-17 25701259-5 2016 In addition, FGF2 significantly prevented sunitinib-induced cardiotoxicity in cardiomyoblast H9c2 cells, possibly via activating the PLC-gamma/c-Raf/CREB pathway. Sunitinib 42-51 Raf-1 proto-oncogene, serine/threonine kinase Rattus norvegicus 143-148 25701259-5 2016 In addition, FGF2 significantly prevented sunitinib-induced cardiotoxicity in cardiomyoblast H9c2 cells, possibly via activating the PLC-gamma/c-Raf/CREB pathway. Sunitinib 42-51 cAMP responsive element binding protein 1 Rattus norvegicus 149-153 25701259-7 2016 Molecular docking simulations further revealed an interaction between the tyrosine kinase domain of FGF receptor 1 (FGFR1) and sunitinib. Sunitinib 127-136 Fibroblast growth factor receptor 1 Rattus norvegicus 100-114 25701259-7 2016 Molecular docking simulations further revealed an interaction between the tyrosine kinase domain of FGF receptor 1 (FGFR1) and sunitinib. Sunitinib 127-136 Fibroblast growth factor receptor 1 Rattus norvegicus 116-121 25701259-8 2016 Taken together, our results clearly demonstrated that FGF2 inhibition plays an important role in sunitinib-induced cardiotoxicity both in vitro and in vivo. Sunitinib 97-106 fibroblast growth factor 2 Rattus norvegicus 54-58 25864572-10 2016 CONCLUSION: Topical administration of both everolimus and sunitinib reduced VEGFR-2 levels and inhibited CNV. Sunitinib 58-67 kinase insert domain receptor Rattus norvegicus 76-83 26779618-3 2016 The antitumor effect of second-line agents is correlated with the type of secondary mutation: indeed, sunitinib is effective against tumors with C-KIT exon 13 or 14 mutations. Sunitinib 102-111 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 145-150 27990160-13 2016 Sunitinib inhibited phospho-ERK1/2 and phospho-mTOR. Sunitinib 0-9 mitogen-activated protein kinase 3 Homo sapiens 28-34 27990160-13 2016 Sunitinib inhibited phospho-ERK1/2 and phospho-mTOR. Sunitinib 0-9 mechanistic target of rapamycin kinase Homo sapiens 47-51 27072203-4 2016 We here review the normal physiology of vascular endothelial growth factor in the kidney, and discuss the pathogenesis, clinical and laboratory manifestations, pathological changes in the kidney, and the management of this uncommon complication of sunitinib. Sunitinib 248-257 vascular endothelial growth factor A Homo sapiens 40-74 26617188-1 2016 Over the last few years, the most recent advances of the molecular mechanisms involved in renal cell carcinoma have led to the use of new drugs targeting VEGF, such as bevacizumab plus interferon, sorafenib, sunitinib, pazopanib, and axitinib, or the mTOR, such as temsirolimus and everolimus. Sunitinib 208-217 vascular endothelial growth factor A Homo sapiens 154-158 26262503-0 2016 Oral Platelet-Derived Growth Factor and Vascular Endothelial Growth Factor Inhibitor Sunitinib Prevents Chronic Allograft Injury in Experimental Kidney Transplantation Model. Sunitinib 85-94 vascular endothelial growth factor A Rattus norvegicus 40-74 26262503-2 2016 Sunitinib is a tyrosine kinase inhibitor which inhibits both VEGF and PDGF receptors. Sunitinib 0-9 vascular endothelial growth factor A Rattus norvegicus 61-65 26262503-12 2016 Sunitinib also inhibited chronic PDGF-A and -B and VEGF-A and -B expressions. Sunitinib 0-9 platelet derived growth factor subunit A Rattus norvegicus 33-46 26385865-5 2016 Furthermore, knockdown of the human ortholog POLG of S. pombe POG1 in human cells significantly increased the cytotoxicity of sunitinib. Sunitinib 126-135 DNA polymerase gamma, catalytic subunit Homo sapiens 45-49 26385865-6 2016 Notably, sunitinib dramatically decreased the levels of POLG mRNAs and proteins, of which downregulation was already known to induce mitochondrial damage of cardiomyocytes, causing cardiotoxicity. Sunitinib 9-18 DNA polymerase gamma, catalytic subunit Homo sapiens 56-60 26385865-7 2016 These results indicate that POLG might play a crucial role in mitochondrial damage as a gene of which expressional pathway is targeted by sunitinib for cardiotoxicity, and that genome-wide drug target screening with GPScreen can be applied to drug toxicity target discovery to understand the molecular insights regarding drug toxicity. Sunitinib 138-147 DNA polymerase gamma, catalytic subunit Homo sapiens 28-32 26262503-12 2016 Sunitinib also inhibited chronic PDGF-A and -B and VEGF-A and -B expressions. Sunitinib 0-9 vascular endothelial growth factor A Rattus norvegicus 51-64 26262503-13 2016 CONCLUSIONS: These results demonstrate that combined inhibition of PGDF and VEGF with sunitinib prevents chronic rejection changes in experimental kidney transplantation which indicates that sunitinib could be a potential intervention also in clinical kidney transplantation. Sunitinib 191-200 vascular endothelial growth factor A Rattus norvegicus 76-80 30695560-1 2016 Sunitinib is one of first targeted agents and tyrosine kinase inhibitors of vascular endothelial growth factor receptor (VEGFR) that approved for therapy of metastatic renal cell carcinoma. Sunitinib 0-9 kinase insert domain receptor Homo sapiens 76-119 30695560-1 2016 Sunitinib is one of first targeted agents and tyrosine kinase inhibitors of vascular endothelial growth factor receptor (VEGFR) that approved for therapy of metastatic renal cell carcinoma. Sunitinib 0-9 kinase insert domain receptor Homo sapiens 121-126 26387812-0 2015 Association of single nucleotide polymorphisms in IL8 and IL13 with sunitinib-induced toxicity in patients with metastatic renal cell carcinoma. Sunitinib 68-77 C-X-C motif chemokine ligand 8 Homo sapiens 50-53 25971960-0 2015 Receptor Tyrosine Kinase Expression Predicts Response to Sunitinib in Breast Cancer. Sunitinib 57-66 ret proto-oncogene Homo sapiens 0-24 25971960-11 2015 Both ERalpha-positive and low-EGFR-expressing tumors had an increased in vitro sunitinib response, as determined by alteration of Erk activation. Sunitinib 79-88 estrogen receptor 1 Homo sapiens 5-12 25971960-11 2015 Both ERalpha-positive and low-EGFR-expressing tumors had an increased in vitro sunitinib response, as determined by alteration of Erk activation. Sunitinib 79-88 epidermal growth factor receptor Homo sapiens 30-34 25971960-11 2015 Both ERalpha-positive and low-EGFR-expressing tumors had an increased in vitro sunitinib response, as determined by alteration of Erk activation. Sunitinib 79-88 EPH receptor B2 Homo sapiens 130-133 26575424-9 2015 Moreover, formononetin enhanced the effect of VEGFR2 inhibitor sunitinib on tumor growth inhibition. Sunitinib 63-72 kinase insert domain receptor Rattus norvegicus 46-52 26484875-2 2015 Clinical trials enrolling ACC patients with high tissue vascular endothelial growth factor receptor (VEGFR) expression levels showed controversial results after treatment with Sunitinib, possibly due to variability in the expression of drug targets, which include epidermal growth factor receptor (EGFR). Sunitinib 176-185 kinase insert domain receptor Homo sapiens 56-99 26484875-2 2015 Clinical trials enrolling ACC patients with high tissue vascular endothelial growth factor receptor (VEGFR) expression levels showed controversial results after treatment with Sunitinib, possibly due to variability in the expression of drug targets, which include epidermal growth factor receptor (EGFR). Sunitinib 176-185 kinase insert domain receptor Homo sapiens 101-106 26484875-2 2015 Clinical trials enrolling ACC patients with high tissue vascular endothelial growth factor receptor (VEGFR) expression levels showed controversial results after treatment with Sunitinib, possibly due to variability in the expression of drug targets, which include epidermal growth factor receptor (EGFR). Sunitinib 176-185 epidermal growth factor receptor Homo sapiens 264-296 26484875-2 2015 Clinical trials enrolling ACC patients with high tissue vascular endothelial growth factor receptor (VEGFR) expression levels showed controversial results after treatment with Sunitinib, possibly due to variability in the expression of drug targets, which include epidermal growth factor receptor (EGFR). Sunitinib 176-185 epidermal growth factor receptor Homo sapiens 102-106 26484875-5 2015 In addition, by cell viability, proliferation and caspase activation assays we found that Sunitinib inhibits adrenocortical cell viability acting, at least in part, through EGFR, that, in turn, is crucial for EGF proliferative effect on adrenocortical cells. Sunitinib 90-99 epidermal growth factor receptor Homo sapiens 173-177 26387812-0 2015 Association of single nucleotide polymorphisms in IL8 and IL13 with sunitinib-induced toxicity in patients with metastatic renal cell carcinoma. Sunitinib 68-77 interleukin 13 Homo sapiens 58-62 26387812-4 2015 METHODS: In this exploratory study, we selected SNPs in genes CYP3A4, NR1I2, POR, IL8, IL13, IL4-R, HIF1A and MET that might possibly be associated with sunitinib treatment outcome. Sunitinib 153-162 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 62-68 26387812-4 2015 METHODS: In this exploratory study, we selected SNPs in genes CYP3A4, NR1I2, POR, IL8, IL13, IL4-R, HIF1A and MET that might possibly be associated with sunitinib treatment outcome. Sunitinib 153-162 nuclear receptor subfamily 1 group I member 2 Homo sapiens 70-75 26387812-4 2015 METHODS: In this exploratory study, we selected SNPs in genes CYP3A4, NR1I2, POR, IL8, IL13, IL4-R, HIF1A and MET that might possibly be associated with sunitinib treatment outcome. Sunitinib 153-162 cytochrome p450 oxidoreductase Homo sapiens 77-80 26387812-4 2015 METHODS: In this exploratory study, we selected SNPs in genes CYP3A4, NR1I2, POR, IL8, IL13, IL4-R, HIF1A and MET that might possibly be associated with sunitinib treatment outcome. Sunitinib 153-162 C-X-C motif chemokine ligand 8 Homo sapiens 82-85 26387812-4 2015 METHODS: In this exploratory study, we selected SNPs in genes CYP3A4, NR1I2, POR, IL8, IL13, IL4-R, HIF1A and MET that might possibly be associated with sunitinib treatment outcome. Sunitinib 153-162 interleukin 13 Homo sapiens 87-91 26387812-4 2015 METHODS: In this exploratory study, we selected SNPs in genes CYP3A4, NR1I2, POR, IL8, IL13, IL4-R, HIF1A and MET that might possibly be associated with sunitinib treatment outcome. Sunitinib 153-162 interleukin 4 receptor Homo sapiens 93-98 26387812-4 2015 METHODS: In this exploratory study, we selected SNPs in genes CYP3A4, NR1I2, POR, IL8, IL13, IL4-R, HIF1A and MET that might possibly be associated with sunitinib treatment outcome. Sunitinib 153-162 hypoxia inducible factor 1 subunit alpha Homo sapiens 100-105 26387812-10 2015 CONCLUSIONS: We show that polymorphisms in IL8 rs1126647 and IL13 rs1800925 are associated with sunitinib-induced toxicities. Sunitinib 96-105 C-X-C motif chemokine ligand 8 Homo sapiens 43-46 26387812-10 2015 CONCLUSIONS: We show that polymorphisms in IL8 rs1126647 and IL13 rs1800925 are associated with sunitinib-induced toxicities. Sunitinib 96-105 interleukin 13 Homo sapiens 61-65 26555641-0 2015 Spectroscopic and molecular docking studies of binding interaction of gefitinib, lapatinib and sunitinib with bovine serum albumin (BSA). Sunitinib 95-104 albumin Homo sapiens 117-130 26555641-1 2015 The binding interactions of three kinds of tyrosine kinase inhibitors (TKIs), such as gefitinib, lapatinib and sunitinib, with bovine serum albumin (BSA) were studied using ultraviolet spectrophotometry, fluorescence spectroscopy, circular dichroism (CD), Fourier transform infrared spectroscopy (FT-IR) and molecular docking methods. Sunitinib 111-120 albumin Homo sapiens 134-147 26201448-0 2015 miR-221/222 Are Involved in Response to Sunitinib Treatment in Metastatic Renal Cell Carcinoma. Sunitinib 40-49 microRNA 221 Homo sapiens 0-7 26474454-8 2015 In contrast, seven other mutations, including phospholipase C-gamma1 (PLCG1) R707Q mutation, were found only in the metastatic tumor, indicating selection of cells with the resistant genotype under sunitinib pressure. Sunitinib 198-207 phospholipase C gamma 1 Homo sapiens 46-68 26474454-8 2015 In contrast, seven other mutations, including phospholipase C-gamma1 (PLCG1) R707Q mutation, were found only in the metastatic tumor, indicating selection of cells with the resistant genotype under sunitinib pressure. Sunitinib 198-207 phospholipase C gamma 1 Homo sapiens 70-75 26386874-3 2015 VEGF-inhibiting strategies include the use of tyrosine kinase inhibitors (sunitinib, axitinib, pazopanib, and sorafenib) and neutralizing antibodies such as bevacizumab. Sunitinib 74-83 vascular endothelial growth factor A Homo sapiens 0-4 26199386-11 2015 Additional analyses suggested (i) prior bevacizumab therapy to be associated with unusually low baseline [sVEGFR2] and (ii) sunitinib causes measurable changes in CTx. Sunitinib 124-133 cytochrome P450 family 27 subfamily A member 1 Homo sapiens 163-166 26199386-13 2015 Sunitinib caused measurable declines in serum CTx. Sunitinib 0-9 cytochrome P450 family 27 subfamily A member 1 Homo sapiens 46-49 28031890-4 2015 STAT3 is also involved in the response to tyrosine kinase inhibitors sunitinib and axitinib, in patients with metastatic renal cell carcinoma, and to second-generation androgen receptor inhibitor enzalutamide in patients with advanced prostate cancer. Sunitinib 69-78 signal transducer and activator of transcription 3 Homo sapiens 0-5 26306766-0 2015 Induction of epithelial-mesenchymal transition via activation of epidermal growth factor receptor contributes to sunitinib resistance in human renal cell carcinoma cell lines. Sunitinib 113-122 epidermal growth factor receptor Homo sapiens 65-97 26306766-3 2015 We aimed to study the association between sunitinib sensitivity and epithelial-mesenchymal transition (EMT) regulation via epidermal growth factor receptor (EGFR) signaling, which is a mechanism of resistance to anticancer drugs. Sunitinib 42-51 epidermal growth factor receptor Homo sapiens 123-155 26306766-3 2015 We aimed to study the association between sunitinib sensitivity and epithelial-mesenchymal transition (EMT) regulation via epidermal growth factor receptor (EGFR) signaling, which is a mechanism of resistance to anticancer drugs. Sunitinib 42-51 epidermal growth factor receptor Homo sapiens 157-161 26306766-6 2015 After treatment with sunitinib, EGFR phosphorylation increased in 786-O cells, resulting in an increase in the phosphorylation of extracellular signal-regulated kinase, nuclear translocation of beta-catenin, and expression of mesenchymal markers. Sunitinib 21-30 epidermal growth factor receptor Homo sapiens 32-36 26306766-6 2015 After treatment with sunitinib, EGFR phosphorylation increased in 786-O cells, resulting in an increase in the phosphorylation of extracellular signal-regulated kinase, nuclear translocation of beta-catenin, and expression of mesenchymal markers. Sunitinib 21-30 catenin beta 1 Homo sapiens 194-206 26306766-7 2015 These results suggest that sunitinib induced EMT via activation of EGFR in 786-O cells, but not in ACHN and Caki-1 cells. Sunitinib 27-36 epidermal growth factor receptor Homo sapiens 67-71 26108242-1 2015 AIMS AND BACKGROUND: The introduction of agents targeting vascular endothelial growth factor has radically changed the approach to metastatic renal cell carcinoma (mRCC): sunitinib and pazopanib are now the standard first-line therapy in mRCC. Sunitinib 171-180 vascular endothelial growth factor A Homo sapiens 58-92 26306766-8 2015 Caki-1/SN cells, a resistant cell line generated by continuous exposure to sunitinib, displayed increased phosphorylation of EGFR. Sunitinib 75-84 epidermal growth factor receptor Homo sapiens 125-129 26306766-9 2015 Cell viability in the presence of sunitinib was decreased by erlotinib, as the selective inhibitor of EGFR, treatment in 786-O and Caki-1/SN cells. Sunitinib 34-43 epidermal growth factor receptor Homo sapiens 102-106 26306766-10 2015 Similarly, erlotinib suppressed sunitinib-induced EGFR activation and upregulated mesenchymal markers. Sunitinib 32-41 epidermal growth factor receptor Homo sapiens 50-54 26306766-11 2015 Thus, we postulate that resistance to sunitinib in RCC may be associated with EMT caused by activation of EGFR. Sunitinib 38-47 epidermal growth factor receptor Homo sapiens 106-110 25963915-8 2015 The VEGF score was significantly decreased similarly by pazopanib, sunitinib, and sorafenib compared to normal saline (P < .05). Sunitinib 67-76 vascular endothelial growth factor A Rattus norvegicus 4-8 26408725-2 2015 Thirteen patients with temozolomide-refractory recurrent anaplastic or low-grade glioma were treated with sunitinib malate, a small-molecule tyrosine kinase inhibitor of the VEGFR, PDGFR, and KIT receptors, in combination with lomustine. Sunitinib 106-122 kinase insert domain receptor Homo sapiens 174-179 26472187-9 2015 Finally, GRP78 knockdown showed potent suppression of tumor growth and enhanced the antitumor effect of sunitinib in RCC xenografts. Sunitinib 104-113 heat shock protein family A (Hsp70) member 5 Homo sapiens 9-14 26513662-0 2015 Prodrug AST-003 Improves the Therapeutic Index of the Multi-Targeted Tyrosine Kinase Inhibitor Sunitinib. Sunitinib 95-104 solute carrier family 17 member 5 Homo sapiens 8-11 26513662-4 2015 The inactive prodrug AST-003 can be converted to Sunitinib in vitro and in vivo. Sunitinib 49-58 solute carrier family 17 member 5 Homo sapiens 21-24 26408740-12 2015 CONCLUSION: A significantly higher expression of VEGF in CRCC in comparison to healthy parenchyma can predict a better response to sunitinib. Sunitinib 131-140 vascular endothelial growth factor A Homo sapiens 49-53 26451083-3 2015 A better understanding of angiogenesis has led to the investigation of drugs that inhibit the vascular endothelial growth factor (VEGF) pathway including anti-VEGF antibody therapy (eg, bevacizumab), inhibitors of angiogenic receptor tyrosine kinases (eg, sunitinib, sorafenib, apatinib, regorafenib), and inhibitors of vascular endothelial growth factor receptors (VEGFRs) (eg, ramucirumab). Sunitinib 256-265 vascular endothelial growth factor A Homo sapiens 94-128 26126494-1 2015 Vascular endothelial growth factor [VEGF] pathway, which plays a key role in angiogenesis, may be blocked by either extracellular interference with VEGF itself (bevacizumab [BEV] or aflibercept), or intracytoplasmic inhibition of VEGF receptor (pazopanib, nintedanib, cediranid, sunitinib and sorafenib). Sunitinib 279-288 vascular endothelial growth factor A Homo sapiens 0-34 26126494-1 2015 Vascular endothelial growth factor [VEGF] pathway, which plays a key role in angiogenesis, may be blocked by either extracellular interference with VEGF itself (bevacizumab [BEV] or aflibercept), or intracytoplasmic inhibition of VEGF receptor (pazopanib, nintedanib, cediranid, sunitinib and sorafenib). Sunitinib 279-288 vascular endothelial growth factor A Homo sapiens 36-40 26126494-1 2015 Vascular endothelial growth factor [VEGF] pathway, which plays a key role in angiogenesis, may be blocked by either extracellular interference with VEGF itself (bevacizumab [BEV] or aflibercept), or intracytoplasmic inhibition of VEGF receptor (pazopanib, nintedanib, cediranid, sunitinib and sorafenib). Sunitinib 279-288 vascular endothelial growth factor A Homo sapiens 148-152 26126494-1 2015 Vascular endothelial growth factor [VEGF] pathway, which plays a key role in angiogenesis, may be blocked by either extracellular interference with VEGF itself (bevacizumab [BEV] or aflibercept), or intracytoplasmic inhibition of VEGF receptor (pazopanib, nintedanib, cediranid, sunitinib and sorafenib). Sunitinib 279-288 vascular endothelial growth factor A Homo sapiens 148-152 25930089-0 2015 CYP3A5 and ABCB1 polymorphisms as predictors for sunitinib outcome in metastatic renal cell carcinoma. Sunitinib 49-58 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 25930089-0 2015 CYP3A5 and ABCB1 polymorphisms as predictors for sunitinib outcome in metastatic renal cell carcinoma. Sunitinib 49-58 ATP binding cassette subfamily B member 1 Homo sapiens 11-16 25930089-11 2015 CONCLUSIONS: The confirmation of previously reported associations between polymorphisms in CYP3A5 and ABCB1 with sunitinib toxicity and efficacy, respectively, indicates that genotyping of these genetic variants will be useful for guiding sunitinib treatment. Sunitinib 113-122 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 91-97 25930089-11 2015 CONCLUSIONS: The confirmation of previously reported associations between polymorphisms in CYP3A5 and ABCB1 with sunitinib toxicity and efficacy, respectively, indicates that genotyping of these genetic variants will be useful for guiding sunitinib treatment. Sunitinib 113-122 ATP binding cassette subfamily B member 1 Homo sapiens 102-107 25930089-11 2015 CONCLUSIONS: The confirmation of previously reported associations between polymorphisms in CYP3A5 and ABCB1 with sunitinib toxicity and efficacy, respectively, indicates that genotyping of these genetic variants will be useful for guiding sunitinib treatment. Sunitinib 239-248 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 91-97 25930089-11 2015 CONCLUSIONS: The confirmation of previously reported associations between polymorphisms in CYP3A5 and ABCB1 with sunitinib toxicity and efficacy, respectively, indicates that genotyping of these genetic variants will be useful for guiding sunitinib treatment. Sunitinib 239-248 ATP binding cassette subfamily B member 1 Homo sapiens 102-107 26451083-3 2015 A better understanding of angiogenesis has led to the investigation of drugs that inhibit the vascular endothelial growth factor (VEGF) pathway including anti-VEGF antibody therapy (eg, bevacizumab), inhibitors of angiogenic receptor tyrosine kinases (eg, sunitinib, sorafenib, apatinib, regorafenib), and inhibitors of vascular endothelial growth factor receptors (VEGFRs) (eg, ramucirumab). Sunitinib 256-265 vascular endothelial growth factor A Homo sapiens 130-134 26015515-1 2015 PURPOSE: The aim of this study was to investigate the effect of VEGF-targeted therapy (sunitinib) on molecular intratumoral heterogeneity (ITH) in metastatic clear cell renal cancer (mccRCC). Sunitinib 87-96 vascular endothelial growth factor A Homo sapiens 64-68 26396737-0 2015 Response to sunitinib of a gastrointestinal stromal tumor with a rare exon 12 PDGFRA mutation. Sunitinib 12-21 platelet derived growth factor receptor alpha Homo sapiens 78-84 26396737-7 2015 CONCLUSIONS: This is the first report detailing a response to treatment with sunitinib of a gastrointestinal stromal tumor with an uncommon exon 12 PDGFRA mutation. Sunitinib 77-86 platelet derived growth factor receptor alpha Homo sapiens 148-154 25922428-5 2015 Sunitinib treatment resulted in a significant reduction in monocytic MDSC, phosphorylated STAT3, and arginase levels in monocytic MDSC (CD33(+)CD14(+)CD16(+)), and an increase in T-cell proliferative activity in cancer patients. Sunitinib 0-9 signal transducer and activator of transcription 3 Homo sapiens 90-95 25922428-5 2015 Sunitinib treatment resulted in a significant reduction in monocytic MDSC, phosphorylated STAT3, and arginase levels in monocytic MDSC (CD33(+)CD14(+)CD16(+)), and an increase in T-cell proliferative activity in cancer patients. Sunitinib 0-9 CD33 molecule Homo sapiens 136-140 26380584-7 2015 In contrast, in RCCs with FLT1 hypermethylation, proliferation inhibition was counteracted by treatment with an anti-FLT1 peptide and both VEGF-TKIs (sunitinib and axitinib). Sunitinib 150-159 fms related receptor tyrosine kinase 1 Homo sapiens 26-30 26380584-7 2015 In contrast, in RCCs with FLT1 hypermethylation, proliferation inhibition was counteracted by treatment with an anti-FLT1 peptide and both VEGF-TKIs (sunitinib and axitinib). Sunitinib 150-159 vascular endothelial growth factor A Homo sapiens 139-143 26380584-8 2015 Demethylation with sunitinib or axitinib synergistically increased proliferation inhibition in the RCCs exhibiting FLT1 hypermethylation. Sunitinib 19-28 fms related receptor tyrosine kinase 1 Homo sapiens 115-119 26380584-9 2015 Using in vitro FLT1 or KDR knockdown models, decreased proliferation inhibition following anti-FLT1 peptide, sunitinib, and axitinib treatment was observed only in FLT1-knockdown cells. Sunitinib 109-118 fms related receptor tyrosine kinase 1 Homo sapiens 15-19 25922428-5 2015 Sunitinib treatment resulted in a significant reduction in monocytic MDSC, phosphorylated STAT3, and arginase levels in monocytic MDSC (CD33(+)CD14(+)CD16(+)), and an increase in T-cell proliferative activity in cancer patients. Sunitinib 0-9 CD14 molecule Homo sapiens 143-147 26380584-9 2015 Using in vitro FLT1 or KDR knockdown models, decreased proliferation inhibition following anti-FLT1 peptide, sunitinib, and axitinib treatment was observed only in FLT1-knockdown cells. Sunitinib 109-118 kinase insert domain receptor Homo sapiens 23-26 26015515-8 2015 Despite this variability, significant chromosomal and transcript changes to key targets of sunitinib, such as VHL, PBRM1, and CAIX, occurred in the treated samples. Sunitinib 91-100 polybromo 1 Homo sapiens 115-120 26380584-10 2015 In patients with renal cancer who received sunitinib, FLT1 promoter methylation was higher in renal cancer tissues from eight nonresponders (stable or progressive disease assessed by the Response Evaluation Criteria in Solid Tumors) than in cancer tissues from five responders (complete response or partial response). Sunitinib 43-52 fms related receptor tyrosine kinase 1 Homo sapiens 54-58 25922428-5 2015 Sunitinib treatment resulted in a significant reduction in monocytic MDSC, phosphorylated STAT3, and arginase levels in monocytic MDSC (CD33(+)CD14(+)CD16(+)), and an increase in T-cell proliferative activity in cancer patients. Sunitinib 0-9 Fc gamma receptor IIIa Homo sapiens 150-154 25922428-7 2015 SBRT synergized the therapeutic effects of sunitinib, especially as related to decreased numbers of monocytic MDSC, Treg, and B cells, and augmented Tbet expression in primary CD4 and CD8 T cells. Sunitinib 43-52 T-box transcription factor 21 Homo sapiens 149-153 25922428-7 2015 SBRT synergized the therapeutic effects of sunitinib, especially as related to decreased numbers of monocytic MDSC, Treg, and B cells, and augmented Tbet expression in primary CD4 and CD8 T cells. Sunitinib 43-52 CD4 molecule Homo sapiens 176-179 26015515-8 2015 Despite this variability, significant chromosomal and transcript changes to key targets of sunitinib, such as VHL, PBRM1, and CAIX, occurred in the treated samples. Sunitinib 91-100 carbonic anhydrase 9 Homo sapiens 126-130 25922428-7 2015 SBRT synergized the therapeutic effects of sunitinib, especially as related to decreased numbers of monocytic MDSC, Treg, and B cells, and augmented Tbet expression in primary CD4 and CD8 T cells. Sunitinib 43-52 CD8a molecule Homo sapiens 184-187 25922428-9 2015 Most interestingly, the responders, defined by sunitinib-mediated reduction in CD33(+)CD11b(+) myeloid cell populations, tend to exhibit improved progression-free survival and cause-specific survival. Sunitinib 47-56 CD33 molecule Homo sapiens 79-83 25922428-9 2015 Most interestingly, the responders, defined by sunitinib-mediated reduction in CD33(+)CD11b(+) myeloid cell populations, tend to exhibit improved progression-free survival and cause-specific survival. Sunitinib 47-56 integrin subunit alpha M Homo sapiens 86-91 25701807-1 2015 Sunitinib is a multi-targeted receptor tyrosine kinase (RTK) inhibitor that blocks several angiogenesis related pathways. Sunitinib 0-9 EPH receptor A3 Gallus gallus 30-54 26366997-8 2015 CONCLUSIONS: The WX-ETK-model-derived vC was an effective prognostic biomarker for advanced HCC treated with sunitinib. Sunitinib 109-118 BMX non-receptor tyrosine kinase Homo sapiens 20-23 25783986-6 2015 Furthermore, TIM-3 rendered RCC cells with the ability to induce resistance to sunitinib and mTOR inhibitors, the standard regimen for patients with ccRCC, as well as stem cell activities. Sunitinib 79-88 hepatitis A virus cellular receptor 2 Homo sapiens 13-18 26014097-8 2015 Treatment of RCC cell lines and RCC xenografts in immunodeficient mice with sunitinib also increased tumor PD-L1 expression. Sunitinib 76-85 CD274 antigen Mus musculus 107-112 25701807-1 2015 Sunitinib is a multi-targeted receptor tyrosine kinase (RTK) inhibitor that blocks several angiogenesis related pathways. Sunitinib 0-9 EPH receptor A3 Gallus gallus 56-59 26713520-0 2015 [Sunitinib suppresses migration of ovarian cancer cells through negative modulation of TGF-beta-mediated epithelial-mesenchymal transition]. Sunitinib 1-10 transforming growth factor beta 1 Homo sapiens 87-95 26067856-5 2015 RESULTS: Sunitinib, which targets the imatinib-inhibited tyrosine kinases of VEGFR, KIT, PDGFR, and FLT-3, was without effect in COV434 and KGN cell lines. Sunitinib 9-18 kinase insert domain receptor Homo sapiens 77-82 26291023-0 2015 Rapid Response to Sunitinib in a Patient with Lung Adenocarcinoma Harboring KIF5B-RET Fusion Gene. Sunitinib 18-27 kinesin family member 5B Homo sapiens 76-81 26291023-0 2015 Rapid Response to Sunitinib in a Patient with Lung Adenocarcinoma Harboring KIF5B-RET Fusion Gene. Sunitinib 18-27 ret proto-oncogene Homo sapiens 82-85 26713520-1 2015 OBJECTIVE: To investigate the effect of sunitinib on the migration of ovarian cells and its mechanism of the negative regulation TGF-beta mediated of epithelial-mesenchymal transition(EMT) by sunitinib to inhibit ovarian cancer metastasis. Sunitinib 40-49 transforming growth factor beta 1 Homo sapiens 129-137 26713520-1 2015 OBJECTIVE: To investigate the effect of sunitinib on the migration of ovarian cells and its mechanism of the negative regulation TGF-beta mediated of epithelial-mesenchymal transition(EMT) by sunitinib to inhibit ovarian cancer metastasis. Sunitinib 192-201 transforming growth factor beta 1 Homo sapiens 129-137 26713520-3 2015 The effects of sunitinib on TGF-beta-induced E-cadherin expression was assessed by Western-blotting, real time RT-PCR and immunofluorescence assay. Sunitinib 15-24 transforming growth factor beta 1 Homo sapiens 28-36 26713520-3 2015 The effects of sunitinib on TGF-beta-induced E-cadherin expression was assessed by Western-blotting, real time RT-PCR and immunofluorescence assay. Sunitinib 15-24 cadherin 1 Homo sapiens 45-55 26713520-4 2015 The protein levels of Snail and the transcriptional activity of Smad in sunitinib-treated cells were examined by Western-blotting and SBE-luciferase assay. Sunitinib 72-81 snail family transcriptional repressor 1 Homo sapiens 22-27 26713520-6 2015 TGF-beta stimulation reduced E-cadherin protein level, which was attenuated by sunitinib. Sunitinib 79-88 transforming growth factor beta 1 Homo sapiens 0-8 26713520-6 2015 TGF-beta stimulation reduced E-cadherin protein level, which was attenuated by sunitinib. Sunitinib 79-88 cadherin 1 Homo sapiens 29-39 26713520-7 2015 Sunitinib inhibited the up-regulation of Snail protein level induced by TGF-beta treatment. Sunitinib 0-9 snail family transcriptional repressor 1 Homo sapiens 41-46 26713520-7 2015 Sunitinib inhibited the up-regulation of Snail protein level induced by TGF-beta treatment. Sunitinib 0-9 transforming growth factor beta 1 Homo sapiens 72-80 26713520-9 2015 A remarkable increment of transcriptional activity of Smads complexes was observed in SKOV3 cells exposed to TGF-beta, which was significantly prohibited by sunitinib. Sunitinib 157-166 transforming growth factor beta 1 Homo sapiens 109-117 26713520-10 2015 CONCLUSION: Sunitinib can inhibit the migration of SKOV3 cells and attenuate the down-regulation of E-cadherin protein level induced by TGF-beta. Sunitinib 12-21 cadherin 1 Homo sapiens 100-110 26713520-10 2015 CONCLUSION: Sunitinib can inhibit the migration of SKOV3 cells and attenuate the down-regulation of E-cadherin protein level induced by TGF-beta. Sunitinib 12-21 transforming growth factor beta 1 Homo sapiens 136-144 26713520-11 2015 Sunitinib can abolish TGF-beta-induced up-regulation of Snail protein and decrease the transcriptional activity of Smad complexes. Sunitinib 0-9 transforming growth factor beta 1 Homo sapiens 22-30 26713520-11 2015 Sunitinib can abolish TGF-beta-induced up-regulation of Snail protein and decrease the transcriptional activity of Smad complexes. Sunitinib 0-9 snail family transcriptional repressor 1 Homo sapiens 56-61 26713520-12 2015 The results indicate that sunitinib suppresses migration of ovarian cancer cells through negative modulation of TGF-beta-mediated epithelial-mesenchymal transition. Sunitinib 26-35 transforming growth factor beta 1 Homo sapiens 112-120 25893276-8 2015 RESULTS: Sunitinib strongly reduced proliferation of PC-3 and DU-145 cells in a dose dependent manner, and decreased levels of p-Akt, p-Erk1/2, and Id-1, compared to untreated cells. Sunitinib 9-18 mitogen-activated protein kinase 3 Homo sapiens 136-142 25963382-6 2015 Tyrosine kinase inhibitors and mammalian target of rapamycin inhibitors are now established classes of drugs used in the treatment of renal cancer, with a total of six having received regulatory approval to date (sorafenib, sunitinib, pazopanib, axitinib, temsirolimus, and everolimus). Sunitinib 224-233 mechanistic target of rapamycin kinase Homo sapiens 31-60 26480622-9 2015 The biologic therapies for metastatic renal cell carcinoma belong to two main groups: angiogenesis inhibitors (VEGF-R inhibitors like sunitinib, sorafenib, pazopanib and axitinib), and inhibitors of the mTOR protein (everolimus and temsirolimus). Sunitinib 134-143 vascular endothelial growth factor A Homo sapiens 111-115 24727344-1 2015 BACKGROUND: Vascular endothelial growth factor inhibitors such as bevacizumab, sorafenib, and sunitinib are utilized in the treatment of multiple cancers. Sunitinib 94-103 vascular endothelial growth factor A Homo sapiens 12-46 26309414-0 2015 Heme oxygenase-1 is a predictive biomarker for therapeutic targeting of advanced clear cell renal cell carcinoma treated with sorafenib or sunitinib. Sunitinib 139-148 heme oxygenase 1 Homo sapiens 0-16 26309414-1 2015 BACKGROUND: We analyzed the expression of heme oxygenase-1 (HO-1) in patients undergoing radical nephrectomy for advanced clear cell renal cell carcinoma (CC-RCC) and evaluated the effects of the targeted therapies treated with sorafenib and sunitinib. Sunitinib 242-251 heme oxygenase 1 Homo sapiens 42-58 26309414-1 2015 BACKGROUND: We analyzed the expression of heme oxygenase-1 (HO-1) in patients undergoing radical nephrectomy for advanced clear cell renal cell carcinoma (CC-RCC) and evaluated the effects of the targeted therapies treated with sorafenib and sunitinib. Sunitinib 242-251 heme oxygenase 1 Homo sapiens 60-64 26309414-6 2015 In the low HO-1 level group, a higher tumor response rate and a longer survival time was achieved in patients who received sorafenib or sunitinib. Sunitinib 136-145 heme oxygenase 1 Homo sapiens 11-15 26309414-9 2015 Overall, HO-1 expression might be a useful biomarker for predicting the response to sunitinib and sorafenib for patients with metastatic CC-RCC. Sunitinib 84-93 heme oxygenase 1 Homo sapiens 9-13 26244574-0 2015 Association of ABCB1 and FLT3 Polymorphisms with Toxicities and Survival in Asian Patients Receiving Sunitinib for Renal Cell Carcinoma. Sunitinib 101-110 ATP binding cassette subfamily B member 1 Homo sapiens 15-20 26244574-0 2015 Association of ABCB1 and FLT3 Polymorphisms with Toxicities and Survival in Asian Patients Receiving Sunitinib for Renal Cell Carcinoma. Sunitinib 101-110 fms related receptor tyrosine kinase 3 Homo sapiens 25-29 26244574-9 2015 In conclusion, ABCB1 and FLT3 polymorphisms may be helpful in predicting sunitinib toxicities, response and survival benefit in Asian mRCC patients. Sunitinib 73-82 ATP binding cassette subfamily B member 1 Homo sapiens 15-20 26244574-9 2015 In conclusion, ABCB1 and FLT3 polymorphisms may be helpful in predicting sunitinib toxicities, response and survival benefit in Asian mRCC patients. Sunitinib 73-82 fms related receptor tyrosine kinase 3 Homo sapiens 25-29 26244574-10 2015 We have also validated the association between FLT3 738T and sunitinib-induced leucopenia previously reported in Caucasian populations, but have not validated other reported genetic associations. Sunitinib 61-70 fms related receptor tyrosine kinase 3 Homo sapiens 47-51 26011058-1 2015 Tyrosine kinase inhibitor sunitinib (used in GIST, advanced RCC, and pancreatic neuroendocrine tumors) undergoes CYP3A4 metabolism and is an ABCB1B and ABCG2 efflux transporters substrate. Sunitinib 26-35 ATP-binding cassette, sub-family B (MDR/TAP), member 1B Mus musculus 141-147 26011058-1 2015 Tyrosine kinase inhibitor sunitinib (used in GIST, advanced RCC, and pancreatic neuroendocrine tumors) undergoes CYP3A4 metabolism and is an ABCB1B and ABCG2 efflux transporters substrate. Sunitinib 26-35 ATP binding cassette subfamily G member 2 (Junior blood group) Mus musculus 152-157 25893276-8 2015 RESULTS: Sunitinib strongly reduced proliferation of PC-3 and DU-145 cells in a dose dependent manner, and decreased levels of p-Akt, p-Erk1/2, and Id-1, compared to untreated cells. Sunitinib 9-18 inhibitor of DNA binding 1, HLH protein Homo sapiens 148-152 28162293-3 2015 VEGF-inhibiting strategies include the use of tyrosine kinase inhibitors (sunitinib, axitinib, pazopanib, and sorafenib) and neutralizing antibodies such as bevacizumab. Sunitinib 74-83 vascular endothelial growth factor A Homo sapiens 0-4 25893276-10 2015 Depletion of Hif-1alpha levels in vitro and a decrease in FMISO uptake in vivo showed that Sunitinib inhibits tumor hypoxia. Sunitinib 91-100 hypoxia inducible factor 1 subunit alpha Homo sapiens 13-23 26151457-2 2015 We investigated the efficacy of sunitinib, a multikinase VEGF inhibitor, in patients with relapsed/refractory GE/oesophageal cancer. Sunitinib 32-41 vascular endothelial growth factor A Homo sapiens 57-61 26198000-1 2015 BACKGROUND: Clinical studies implying the sunitinib multi-kinase inhibitor have led to disappointing results for breast cancer care but mostly focused on HER2-negative subtypes. Sunitinib 42-51 erb-b2 receptor tyrosine kinase 2 Mus musculus 154-158 26198000-13 2015 CONCLUSIONS: [(18)F]FDG and [(18)F]FMISO PET were relevant approaches to study the response to sunitinib in this luminal B (HER2-positive) model. Sunitinib 95-104 erb-b2 receptor tyrosine kinase 2 Mus musculus 124-128 25980323-1 2015 The aim of this study was to evaluate the capacity of BR55, an ultrasound contrast agent specifically targeting vascular endothelial growth factor receptor 2 (VEGFR2), to distinguish the specific anti-VEGFR2 therapy effect of sunitinib from other anti-angiogenic effects of a therapy (imatinib) that does not directly inhibit VEGFR2. Sunitinib 226-235 kinase insert domain protein receptor Mus musculus 112-157 25980323-1 2015 The aim of this study was to evaluate the capacity of BR55, an ultrasound contrast agent specifically targeting vascular endothelial growth factor receptor 2 (VEGFR2), to distinguish the specific anti-VEGFR2 therapy effect of sunitinib from other anti-angiogenic effects of a therapy (imatinib) that does not directly inhibit VEGFR2. Sunitinib 226-235 kinase insert domain protein receptor Mus musculus 159-165 25980323-1 2015 The aim of this study was to evaluate the capacity of BR55, an ultrasound contrast agent specifically targeting vascular endothelial growth factor receptor 2 (VEGFR2), to distinguish the specific anti-VEGFR2 therapy effect of sunitinib from other anti-angiogenic effects of a therapy (imatinib) that does not directly inhibit VEGFR2. Sunitinib 226-235 kinase insert domain protein receptor Mus musculus 201-207 25980323-1 2015 The aim of this study was to evaluate the capacity of BR55, an ultrasound contrast agent specifically targeting vascular endothelial growth factor receptor 2 (VEGFR2), to distinguish the specific anti-VEGFR2 therapy effect of sunitinib from other anti-angiogenic effects of a therapy (imatinib) that does not directly inhibit VEGFR2. Sunitinib 226-235 kinase insert domain protein receptor Mus musculus 201-207 25980323-9 2015 Targeted contrast-enhanced imaging revealed lower differential targeted enhancement, that is, lower levels of VEGFR2 expression, in sunitinib-treated mice relative to placebo-treated mice from 24 h (p < 0.05) and relative to both placebo- and imatinib-treated mice from 48 h (p < 0.05). Sunitinib 132-141 kinase insert domain protein receptor Mus musculus 110-116 26009164-3 2015 Results reveal that some compounds have potent antitumor activity, and the most active 13c7 (IC50s: 4.49-15.39 muM) was found to be more active than Sunitinib (IC50s: 4.70->30 muM) against all of the tested cancer cell lines. Sunitinib 149-158 latexin Homo sapiens 179-182 26151457-18 2015 On this study, patients with clinical benefit from sunitinib had higher tumour CFB expression, and thus has identified CFB as a potential predictor for efficacy of anti-angiogenic therapy. Sunitinib 51-60 complement factor B Homo sapiens 79-82 26114873-0 2015 Role of IL13RA2 in Sunitinib Resistance in Clear Cell Renal Cell Carcinoma. Sunitinib 19-28 interleukin 13 receptor subunit alpha 2 Homo sapiens 8-15 26114873-2 2015 Sunitinib is an agent that targets VEGF receptors and is considered to be a standard treatment for metastatic or unresectable clear cell RCC (ccRCC). Sunitinib 0-9 vascular endothelial growth factor A Homo sapiens 35-39 26114873-7 2015 Comparing gene expression profiles between the two xenograft models with different sensitivity to sunitinib, we identified interleukin 13 receptor alpha 2 (IL13RA2) as a candidate molecule associated with the acquired sunitinib-resistance in ccRCC. Sunitinib 98-107 interleukin 13 receptor subunit alpha 2 Homo sapiens 123-154 26114873-7 2015 Comparing gene expression profiles between the two xenograft models with different sensitivity to sunitinib, we identified interleukin 13 receptor alpha 2 (IL13RA2) as a candidate molecule associated with the acquired sunitinib-resistance in ccRCC. Sunitinib 98-107 interleukin 13 receptor subunit alpha 2 Homo sapiens 156-163 26114873-7 2015 Comparing gene expression profiles between the two xenograft models with different sensitivity to sunitinib, we identified interleukin 13 receptor alpha 2 (IL13RA2) as a candidate molecule associated with the acquired sunitinib-resistance in ccRCC. Sunitinib 218-227 interleukin 13 receptor subunit alpha 2 Homo sapiens 123-154 26114873-7 2015 Comparing gene expression profiles between the two xenograft models with different sensitivity to sunitinib, we identified interleukin 13 receptor alpha 2 (IL13RA2) as a candidate molecule associated with the acquired sunitinib-resistance in ccRCC. Sunitinib 218-227 interleukin 13 receptor subunit alpha 2 Homo sapiens 156-163 26114873-8 2015 And patients with high IL13RA2 expression in immunohistochemistry in primary ccRCC tumor tends to have sunitinib-resistant metastatic site. Sunitinib 103-112 interleukin 13 receptor subunit alpha 2 Homo sapiens 23-30 26114873-9 2015 Next, we showed that sunitinib-sensitive 786-O cells acquired resistance in vivo when IL13RA2 was overexpressed. Sunitinib 21-30 interleukin 13 receptor subunit alpha 2 Homo sapiens 86-93 26114873-10 2015 Conversely, shRNA-mediated knockdown of IL13RA2 successfully overcame the sunitinib-resistance in Caki-1 cells. Sunitinib 74-83 interleukin 13 receptor subunit alpha 2 Homo sapiens 40-47 26114873-11 2015 Histopathological analyses revealed that IL13RA2 repressed sunitinib-induced apoptosis without increasing tumor vasculature in vivo. Sunitinib 59-68 interleukin 13 receptor subunit alpha 2 Homo sapiens 41-48 26114873-12 2015 To our knowledge, this is a novel mechanism of developing resistance to sunitinib in a certain population of ccRCC, and these results indicate that IL13RA2 could be one of potential target to overcome sunitinib resistance. Sunitinib 72-81 interleukin 13 receptor subunit alpha 2 Homo sapiens 148-155 26114873-12 2015 To our knowledge, this is a novel mechanism of developing resistance to sunitinib in a certain population of ccRCC, and these results indicate that IL13RA2 could be one of potential target to overcome sunitinib resistance. Sunitinib 201-210 interleukin 13 receptor subunit alpha 2 Homo sapiens 148-155 26070816-9 2015 In the melanoma xenograft model, serum phospho-CSE1L level declined 5 days after vemurafenib/sunitinib treatment and 3 days after sorafenib/lapatinib treatment in the HT-29 colon cancer xenograft model. Sunitinib 93-102 chromosome segregation 1-like (S. cerevisiae) Mus musculus 47-52 25564360-0 2015 Synthetic miR-145 Mimic Enhances the Cytotoxic Effect of the Antiangiogenic Drug Sunitinib in Glioblastoma. Sunitinib 81-90 microRNA 145 Homo sapiens 10-17 25861727-0 2015 MiRNA-21 silencing mediated by tumor-targeted nanoparticles combined with sunitinib: A new multimodal gene therapy approach for glioblastoma. Sunitinib 74-83 microRNA 21 Homo sapiens 0-8 25861727-5 2015 Decreased tumor cell proliferation and tumor size, as well as enhanced apoptosis activation and, to a lesser extent, improvement of animal survival, were also observed in GBM-bearing mice upon systemic delivery of targeted nanoparticle-formulated anti-miR-21 oligonucleotides and exposure to the tyrosine kinase inhibitor sunitinib. Sunitinib 322-331 microRNA 21a Mus musculus 252-258 25818407-0 2015 A phase I/II study of sunitinib and intensive chemotherapy in patients over 60 years of age with acute myeloid leukaemia and activating FLT3 mutations. Sunitinib 22-31 fms related receptor tyrosine kinase 3 Homo sapiens 136-140 26070816-0 2015 Early decline in serum phospho-CSE1L levels in vemurafenib/sunitinib-treated melanoma and sorafenib/lapatinib-treated colorectal tumor xenografts. Sunitinib 59-68 chromosome segregation 1-like (S. cerevisiae) Mus musculus 31-36 26042424-1 2015 Inhibitors of VEGF receptor (VEGFR) signaling such as sorafenib and sunitinib that are currently used in the treatment of malignant diseases have been shown to affect immunological responses by inhibition of the function of antigen presenting cells and T lymphocytes. Sunitinib 68-77 kinase insert domain receptor Homo sapiens 14-27 26042424-1 2015 Inhibitors of VEGF receptor (VEGFR) signaling such as sorafenib and sunitinib that are currently used in the treatment of malignant diseases have been shown to affect immunological responses by inhibition of the function of antigen presenting cells and T lymphocytes. Sunitinib 68-77 kinase insert domain receptor Homo sapiens 29-34 25564360-4 2015 In addition, we suggested that the enhanced cytotoxicity of Sunitinib by miR-145 mimic was mediated by inhibition of both P-gp and Bcrp. Sunitinib 60-69 microRNA 145 Homo sapiens 73-80 25564360-4 2015 In addition, we suggested that the enhanced cytotoxicity of Sunitinib by miR-145 mimic was mediated by inhibition of both P-gp and Bcrp. Sunitinib 60-69 phosphoglycolate phosphatase Homo sapiens 122-126 25564360-4 2015 In addition, we suggested that the enhanced cytotoxicity of Sunitinib by miR-145 mimic was mediated by inhibition of both P-gp and Bcrp. Sunitinib 60-69 BCR pseudogene 1 Homo sapiens 131-135 25488966-2 2015 Recent placebo-controlled phase III trials of the mammalian target of rapamycin (mTOR) inhibitor everolimus and the vascular endothelial growth factor (VEGF)/platelet-derived growth factor receptor inhibitor sunitinib have noted improved progression-free survival (PFS). Sunitinib 208-217 vascular endothelial growth factor A Homo sapiens 116-150 25917740-5 2015 However, compared with PC3/C, PC3/sh-Akt1 showed a significantly higher sensitivity to sunitinib, accompanying impaired phosphorylation of p44/42 mitogen-activated protein kinase, downregulation of Bcl-2, and upregulation of Bax. Sunitinib 87-96 proprotein convertase subtilisin/kexin type 1 Homo sapiens 30-33 25948774-5 2015 At variance, only VEGF-receptor inhibition using the non-specific tyrosine kinase inhibitor Sunitinib or the anti-VEGF-receptor 2 neutralizing antibody, but not VEGF blockade using Bevacizumab, impaired the process of endothelial differentiation in vitro, suggesting a VEGF-independent mechanism. Sunitinib 92-101 vascular endothelial growth factor A Mus musculus 18-22 25907705-4 2015 Its potent and selective inhibition of VEGFR was the rationale for its development in the second-line setting after failure of prior cytokines or sunitinib. Sunitinib 146-155 kinase insert domain receptor Homo sapiens 39-44 26003083-0 2015 Effect of enhanced expression of connexin 43 on sunitinib-induced cytotoxicity in mesothelioma cells. Sunitinib 48-57 gap junction protein alpha 1 Homo sapiens 33-44 26003083-4 2015 This work aims to evaluate the effect of the restoration of connexin 43 (Cx43) (the most ubiquitous Cx subtype) expression on sunitinib (SU)-induced cytotoxicity in malignant mesothelioma (MM) cells. Sunitinib 126-135 gap junction protein alpha 1 Homo sapiens 60-71 26003083-4 2015 This work aims to evaluate the effect of the restoration of connexin 43 (Cx43) (the most ubiquitous Cx subtype) expression on sunitinib (SU)-induced cytotoxicity in malignant mesothelioma (MM) cells. Sunitinib 126-135 gap junction protein alpha 1 Homo sapiens 73-77 26003083-4 2015 This work aims to evaluate the effect of the restoration of connexin 43 (Cx43) (the most ubiquitous Cx subtype) expression on sunitinib (SU)-induced cytotoxicity in malignant mesothelioma (MM) cells. Sunitinib 126-135 LOC100128922 Homo sapiens 73-75 26003083-4 2015 This work aims to evaluate the effect of the restoration of connexin 43 (Cx43) (the most ubiquitous Cx subtype) expression on sunitinib (SU)-induced cytotoxicity in malignant mesothelioma (MM) cells. Sunitinib 137-139 gap junction protein alpha 1 Homo sapiens 60-71 26003083-4 2015 This work aims to evaluate the effect of the restoration of connexin 43 (Cx43) (the most ubiquitous Cx subtype) expression on sunitinib (SU)-induced cytotoxicity in malignant mesothelioma (MM) cells. Sunitinib 137-139 gap junction protein alpha 1 Homo sapiens 73-77 26003083-4 2015 This work aims to evaluate the effect of the restoration of connexin 43 (Cx43) (the most ubiquitous Cx subtype) expression on sunitinib (SU)-induced cytotoxicity in malignant mesothelioma (MM) cells. Sunitinib 137-139 LOC100128922 Homo sapiens 73-75 26003083-5 2015 Increased Cx43 expression in an MM cell line (H28) improved the ability of SU to inhibit receptor tyrosine kinase (RTK) signaling. Sunitinib 75-77 gap junction protein alpha 1 Homo sapiens 10-14 26003083-5 2015 Increased Cx43 expression in an MM cell line (H28) improved the ability of SU to inhibit receptor tyrosine kinase (RTK) signaling. Sunitinib 75-77 ret proto-oncogene Homo sapiens 89-113 26003083-5 2015 Increased Cx43 expression in an MM cell line (H28) improved the ability of SU to inhibit receptor tyrosine kinase (RTK) signaling. Sunitinib 75-77 ret proto-oncogene Homo sapiens 115-118 26003083-6 2015 Moreover, higher Cx43 expression promoted SU-induced apoptosis. Sunitinib 42-44 gap junction protein alpha 1 Homo sapiens 17-21 26003083-7 2015 The cell viability test revealed that Cx43 enhanced the cytotoxic effect of SU in a GJ-independent manner. Sunitinib 76-78 gap junction protein alpha 1 Homo sapiens 38-42 26003083-10 2015 Furthermore, higher Cx43 expression increased the production of a cleaved (active) form of Bax during SU-induced apoptosis with no alteration in total Bax expression. Sunitinib 102-104 gap junction protein alpha 1 Homo sapiens 20-24 26003083-10 2015 Furthermore, higher Cx43 expression increased the production of a cleaved (active) form of Bax during SU-induced apoptosis with no alteration in total Bax expression. Sunitinib 102-104 BCL2 associated X, apoptosis regulator Homo sapiens 91-94 26003083-11 2015 These findings indicate that Cx43 most likely increases sensitivity to SU in H28 through direct interaction with Bax. Sunitinib 71-73 gap junction protein alpha 1 Homo sapiens 29-33 26003083-11 2015 These findings indicate that Cx43 most likely increases sensitivity to SU in H28 through direct interaction with Bax. Sunitinib 71-73 BCL2 associated X, apoptosis regulator Homo sapiens 113-116 25917740-0 2015 Enhanced Sensitivity to Sunitinib by Inhibition of Akt1 Expression in Human Castration-resistant Prostate Cancer PC3 Cells Both In Vitro and In Vivo. Sunitinib 24-33 AKT serine/threonine kinase 1 Homo sapiens 51-55 25917740-0 2015 Enhanced Sensitivity to Sunitinib by Inhibition of Akt1 Expression in Human Castration-resistant Prostate Cancer PC3 Cells Both In Vitro and In Vivo. Sunitinib 24-33 proprotein convertase subtilisin/kexin type 1 Homo sapiens 113-116 25917740-1 2015 OBJECTIVE: To investigate whether antitumor activity of sunitinib is enhanced by silencing Akt1 in a human castration-resistant prostate cancer PC3 model. Sunitinib 56-65 AKT serine/threonine kinase 1 Homo sapiens 91-95 25917740-3 2015 Changes in various phenotypes of PC3/sh-Akt1 after treatment with sunitinib were compared with those of PC3 transfected with control vector alone (PC3/C) both in vitro and in vivo. Sunitinib 66-75 proprotein convertase subtilisin/kexin type 1 Homo sapiens 33-36 25917740-3 2015 Changes in various phenotypes of PC3/sh-Akt1 after treatment with sunitinib were compared with those of PC3 transfected with control vector alone (PC3/C) both in vitro and in vivo. Sunitinib 66-75 AKT serine/threonine kinase 1 Homo sapiens 40-44 25752672-8 2015 Vascular endothelial growth factor-A blocker 1 effectively restored VEGF levels, decreased cerebral oedema, and reduced vascular leakage under hypobaric hypoxia when compared to sunitinib-treated rats. Sunitinib 178-187 vascular endothelial growth factor A Rattus norvegicus 0-36 25917740-5 2015 However, compared with PC3/C, PC3/sh-Akt1 showed a significantly higher sensitivity to sunitinib, accompanying impaired phosphorylation of p44/42 mitogen-activated protein kinase, downregulation of Bcl-2, and upregulation of Bax. Sunitinib 87-96 AKT serine/threonine kinase 1 Homo sapiens 37-41 25917740-6 2015 In addition, treatment with sunitinib significantly suppressed the migration ability of PC3/sh-Akt1 compared with that of PC3/C. Sunitinib 28-37 proprotein convertase subtilisin/kexin type 1 Homo sapiens 88-91 25917740-6 2015 In addition, treatment with sunitinib significantly suppressed the migration ability of PC3/sh-Akt1 compared with that of PC3/C. Sunitinib 28-37 AKT serine/threonine kinase 1 Homo sapiens 95-99 25917740-6 2015 In addition, treatment with sunitinib significantly suppressed the migration ability of PC3/sh-Akt1 compared with that of PC3/C. Sunitinib 28-37 proprotein convertase subtilisin/kexin type 1 Homo sapiens 122-125 25917740-7 2015 In vivo, administration of sunitinib induced the significantly marked growth inhibition of PC3/sh-Akt1 compared with that of PC3/C, and apoptotic index in PC3/sh-Akt1 tumor in mice treated with sunitinib was significantly greater than that in PC3/C tumor. Sunitinib 27-36 proprotein convertase subtilisin/kexin type 1 Mus musculus 91-94 25917740-7 2015 In vivo, administration of sunitinib induced the significantly marked growth inhibition of PC3/sh-Akt1 compared with that of PC3/C, and apoptotic index in PC3/sh-Akt1 tumor in mice treated with sunitinib was significantly greater than that in PC3/C tumor. Sunitinib 27-36 thymoma viral proto-oncogene 1 Mus musculus 98-102 25917740-7 2015 In vivo, administration of sunitinib induced the significantly marked growth inhibition of PC3/sh-Akt1 compared with that of PC3/C, and apoptotic index in PC3/sh-Akt1 tumor in mice treated with sunitinib was significantly greater than that in PC3/C tumor. Sunitinib 27-36 proprotein convertase subtilisin/kexin type 1 Mus musculus 125-128 25917740-7 2015 In vivo, administration of sunitinib induced the significantly marked growth inhibition of PC3/sh-Akt1 compared with that of PC3/C, and apoptotic index in PC3/sh-Akt1 tumor in mice treated with sunitinib was significantly greater than that in PC3/C tumor. Sunitinib 27-36 proprotein convertase subtilisin/kexin type 1 Mus musculus 125-128 25917740-7 2015 In vivo, administration of sunitinib induced the significantly marked growth inhibition of PC3/sh-Akt1 compared with that of PC3/C, and apoptotic index in PC3/sh-Akt1 tumor in mice treated with sunitinib was significantly greater than that in PC3/C tumor. Sunitinib 27-36 thymoma viral proto-oncogene 1 Mus musculus 162-166 25917740-7 2015 In vivo, administration of sunitinib induced the significantly marked growth inhibition of PC3/sh-Akt1 compared with that of PC3/C, and apoptotic index in PC3/sh-Akt1 tumor in mice treated with sunitinib was significantly greater than that in PC3/C tumor. Sunitinib 27-36 proprotein convertase subtilisin/kexin type 1 Mus musculus 125-128 25917740-7 2015 In vivo, administration of sunitinib induced the significantly marked growth inhibition of PC3/sh-Akt1 compared with that of PC3/C, and apoptotic index in PC3/sh-Akt1 tumor in mice treated with sunitinib was significantly greater than that in PC3/C tumor. Sunitinib 194-203 thymoma viral proto-oncogene 1 Mus musculus 98-102 25917740-7 2015 In vivo, administration of sunitinib induced the significantly marked growth inhibition of PC3/sh-Akt1 compared with that of PC3/C, and apoptotic index in PC3/sh-Akt1 tumor in mice treated with sunitinib was significantly greater than that in PC3/C tumor. Sunitinib 194-203 thymoma viral proto-oncogene 1 Mus musculus 162-166 25862847-15 2015 Everolimus, sorafenib and sunitinib significantly reduced mTOR expression. Sunitinib 26-35 mechanistic target of rapamycin kinase Homo sapiens 58-62 25862847-17 2015 Sunitinib and sorafenib increased AREG expression in HNSCC 11A and 14C but not in CERV196. Sunitinib 0-9 amphiregulin Homo sapiens 34-38 25862847-19 2015 Interestingly, sorafenib and sunitinib increased AREG in HNSCC 11A and 14C, which could be a possible evasive mechanism following incubation with these drugs. Sunitinib 29-38 amphiregulin Homo sapiens 49-53 25862847-20 2015 On the contrary, p16-positive CERV196 showed increased susceptibility to sorafenib and sunitinib concerning suppression of AREG expression. Sunitinib 87-96 cyclin dependent kinase inhibitor 2A Homo sapiens 17-20 25862847-20 2015 On the contrary, p16-positive CERV196 showed increased susceptibility to sorafenib and sunitinib concerning suppression of AREG expression. Sunitinib 87-96 amphiregulin Homo sapiens 123-127 25123505-0 2015 Expression of TNF-alpha and CD44 is implicated in poor prognosis, cancer cell invasion, metastasis and resistance to the sunitinib treatment in clear cell renal cell carcinomas. Sunitinib 121-130 tumor necrosis factor Homo sapiens 14-23 25123505-0 2015 Expression of TNF-alpha and CD44 is implicated in poor prognosis, cancer cell invasion, metastasis and resistance to the sunitinib treatment in clear cell renal cell carcinomas. Sunitinib 121-130 CD44 molecule (Indian blood group) Homo sapiens 28-32 25123505-9 2015 Among the 25 ccRCC patients treated with sunitinib for metastatic disease, high CD44 expression was associated with poor treatment outcome. Sunitinib 41-50 CD44 molecule (Indian blood group) Homo sapiens 80-84 25123505-10 2015 Importantly, residual carcinoma cells in the sunitinib-treated metastatic ccRCCs were strongly positive for CD44, and the CD44 expression was significantly higher in the tumors from the sunitinib-treated patients than in those from untreated ones. Sunitinib 45-54 CD44 molecule (Indian blood group) Homo sapiens 108-112 25123505-10 2015 Importantly, residual carcinoma cells in the sunitinib-treated metastatic ccRCCs were strongly positive for CD44, and the CD44 expression was significantly higher in the tumors from the sunitinib-treated patients than in those from untreated ones. Sunitinib 45-54 CD44 molecule (Indian blood group) Homo sapiens 122-126 25123505-10 2015 Importantly, residual carcinoma cells in the sunitinib-treated metastatic ccRCCs were strongly positive for CD44, and the CD44 expression was significantly higher in the tumors from the sunitinib-treated patients than in those from untreated ones. Sunitinib 186-195 CD44 molecule (Indian blood group) Homo sapiens 122-126 26097571-5 2015 Dual luciferase analysis confirmed that Raf/MEK/ERK pathway could directly be regulated by SOX9, and sequential experiments demonstrated that, renal carcinoma cells could sensitize to Sorafenib/Sunitinib through Raf/MEK/ERK signaling pathway inhibition regulated by SOX9 down-regulation. Sunitinib 194-203 zinc fingers and homeoboxes 2 Homo sapiens 212-215 26097571-5 2015 Dual luciferase analysis confirmed that Raf/MEK/ERK pathway could directly be regulated by SOX9, and sequential experiments demonstrated that, renal carcinoma cells could sensitize to Sorafenib/Sunitinib through Raf/MEK/ERK signaling pathway inhibition regulated by SOX9 down-regulation. Sunitinib 194-203 zinc fingers and homeoboxes 2 Homo sapiens 40-43 26097571-5 2015 Dual luciferase analysis confirmed that Raf/MEK/ERK pathway could directly be regulated by SOX9, and sequential experiments demonstrated that, renal carcinoma cells could sensitize to Sorafenib/Sunitinib through Raf/MEK/ERK signaling pathway inhibition regulated by SOX9 down-regulation. Sunitinib 194-203 mitogen-activated protein kinase kinase 7 Homo sapiens 44-47 26097571-5 2015 Dual luciferase analysis confirmed that Raf/MEK/ERK pathway could directly be regulated by SOX9, and sequential experiments demonstrated that, renal carcinoma cells could sensitize to Sorafenib/Sunitinib through Raf/MEK/ERK signaling pathway inhibition regulated by SOX9 down-regulation. Sunitinib 194-203 mitogen-activated protein kinase 1 Homo sapiens 48-51 26097571-5 2015 Dual luciferase analysis confirmed that Raf/MEK/ERK pathway could directly be regulated by SOX9, and sequential experiments demonstrated that, renal carcinoma cells could sensitize to Sorafenib/Sunitinib through Raf/MEK/ERK signaling pathway inhibition regulated by SOX9 down-regulation. Sunitinib 194-203 SRY-box transcription factor 9 Homo sapiens 91-95 26097571-5 2015 Dual luciferase analysis confirmed that Raf/MEK/ERK pathway could directly be regulated by SOX9, and sequential experiments demonstrated that, renal carcinoma cells could sensitize to Sorafenib/Sunitinib through Raf/MEK/ERK signaling pathway inhibition regulated by SOX9 down-regulation. Sunitinib 194-203 mitogen-activated protein kinase kinase 7 Homo sapiens 216-219 26097571-5 2015 Dual luciferase analysis confirmed that Raf/MEK/ERK pathway could directly be regulated by SOX9, and sequential experiments demonstrated that, renal carcinoma cells could sensitize to Sorafenib/Sunitinib through Raf/MEK/ERK signaling pathway inhibition regulated by SOX9 down-regulation. Sunitinib 194-203 mitogen-activated protein kinase 1 Homo sapiens 220-223 26097571-5 2015 Dual luciferase analysis confirmed that Raf/MEK/ERK pathway could directly be regulated by SOX9, and sequential experiments demonstrated that, renal carcinoma cells could sensitize to Sorafenib/Sunitinib through Raf/MEK/ERK signaling pathway inhibition regulated by SOX9 down-regulation. Sunitinib 194-203 SRY-box transcription factor 9 Homo sapiens 266-270 25123505-11 2015 Our data show that TNF-alpha plays an important role in progression of ccRCCs by inducing EMT and CD44 expression, and suggest that CD44 induced by TNF-alpha may be involved in the resistance to the sunitinib treatment. Sunitinib 199-208 tumor necrosis factor Homo sapiens 19-28 26097571-6 2015 In a small cases with mRCC treated with Sorafenib/Sunitinib (n=38), comparative analysis showed that patients with SOX9 (-) had much better therapeutic response to TKIs than those with SOX9 (+) (PD: 9.1% vs. 56.2%, P=0.002, DCR: 90.9% vs. 43.8%, P=0.002). Sunitinib 50-59 SRY-box transcription factor 9 Homo sapiens 115-119 25123505-11 2015 Our data show that TNF-alpha plays an important role in progression of ccRCCs by inducing EMT and CD44 expression, and suggest that CD44 induced by TNF-alpha may be involved in the resistance to the sunitinib treatment. Sunitinib 199-208 CD44 molecule (Indian blood group) Homo sapiens 98-102 25123505-11 2015 Our data show that TNF-alpha plays an important role in progression of ccRCCs by inducing EMT and CD44 expression, and suggest that CD44 induced by TNF-alpha may be involved in the resistance to the sunitinib treatment. Sunitinib 199-208 CD44 molecule (Indian blood group) Homo sapiens 132-136 25123505-11 2015 Our data show that TNF-alpha plays an important role in progression of ccRCCs by inducing EMT and CD44 expression, and suggest that CD44 induced by TNF-alpha may be involved in the resistance to the sunitinib treatment. Sunitinib 199-208 tumor necrosis factor Homo sapiens 148-157 25695485-0 2015 IL8 polymorphisms and overall survival in pazopanib- or sunitinib-treated patients with renal cell carcinoma. Sunitinib 56-65 C-X-C motif chemokine ligand 8 Homo sapiens 0-3 25653444-4 2015 We also discovered that AAK1 and GAK inhibitors, including the approved anticancer drugs sunitinib and erlotinib, could block HCV assembly. Sunitinib 89-98 AP2 associated kinase 1 Homo sapiens 24-28 25653444-4 2015 We also discovered that AAK1 and GAK inhibitors, including the approved anticancer drugs sunitinib and erlotinib, could block HCV assembly. Sunitinib 89-98 cyclin G associated kinase Homo sapiens 33-36 25653444-9 2015 Last, in addition to affecting assembly, sunitinib and erlotinib inhibited HCV entry at a postbinding step, their combination was synergistic, and their antiviral effect was reversed by either AAK1 or GAK overexpression. Sunitinib 41-50 AP2 associated kinase 1 Homo sapiens 193-197 25653444-9 2015 Last, in addition to affecting assembly, sunitinib and erlotinib inhibited HCV entry at a postbinding step, their combination was synergistic, and their antiviral effect was reversed by either AAK1 or GAK overexpression. Sunitinib 41-50 cyclin G associated kinase Homo sapiens 201-204 25653444-10 2015 Together, these results validate AAK1 and GAK as critical regulators of HCV entry that function in part by activating EGFR, AP2M1, and NUMB and as the molecular targets underlying the antiviral effect of sunitinib and erlotinib (in addition to EGFR), respectively. Sunitinib 204-213 AP2 associated kinase 1 Homo sapiens 33-37 25653444-10 2015 Together, these results validate AAK1 and GAK as critical regulators of HCV entry that function in part by activating EGFR, AP2M1, and NUMB and as the molecular targets underlying the antiviral effect of sunitinib and erlotinib (in addition to EGFR), respectively. Sunitinib 204-213 cyclin G associated kinase Homo sapiens 42-45 25756398-10 2015 CONCLUSIONS: Interleukin-8, sVEGFR-3, and SDF-1alpha were identified as predictors of sunitinib clinical outcome. Sunitinib 86-95 C-X-C motif chemokine ligand 8 Homo sapiens 13-26 25695485-7 2015 CONCLUSIONS: Variant alleles of IL8 polymorphisms are associated with poorer survival outcomes in pazopanib- or sunitinib-treated patients with aRCC. Sunitinib 112-121 C-X-C motif chemokine ligand 8 Homo sapiens 32-35 25756398-11 2015 Putative pro-tumorigenic CXCR4+ and VEGFR-1+ monocytes represent novel candidate markers and biologically relevant targets explaining the activity of sunitinib. Sunitinib 150-159 C-X-C motif chemokine receptor 4 Homo sapiens 25-30 25776476-0 2015 Cytotoxic activity of sunitinib and everolimus in Caki-1 renal cancer cells is accompanied by modulations in the expression of apoptosis-related microRNA clusters and BCL2 family genes. Sunitinib 22-31 BCL2 apoptosis regulator Homo sapiens 167-171 25756398-11 2015 Putative pro-tumorigenic CXCR4+ and VEGFR-1+ monocytes represent novel candidate markers and biologically relevant targets explaining the activity of sunitinib. Sunitinib 150-159 fms related receptor tyrosine kinase 1 Homo sapiens 36-43 25849942-4 2015 Upon Cox-2 inhibition PGE2 levels were normalized and efficacy of anti-vascular endothelial growth factor receptor 2 (anti-VEGFR-2) antibodies and sunitinib was enhanced. Sunitinib 147-156 prostaglandin-endoperoxide synthase 2 Homo sapiens 5-10 25750290-7 2015 Sunitinib, sorafenib and everolimus significantly reduced the expression of VEGFR1 and -2, especially in p16-positive CERV196 cells. Sunitinib 0-9 fms related receptor tyrosine kinase 1 Homo sapiens 76-89 25750290-8 2015 Sunitinib appeared to be more effective in reducing VEGFR1 and -2 expression than sorafenib and everolimus. Sunitinib 0-9 fms related receptor tyrosine kinase 1 Homo sapiens 52-65 25750290-10 2015 PTEN expression increased significantly under sunitinib and sorafenib in HNSCC 11A and CERV196 cells. Sunitinib 46-55 phosphatase and tensin homolog Homo sapiens 0-4 25750290-13 2015 Sunitinib and sorafenib were able to increase PTEN expression, which might induce apoptosis of cancer cells. Sunitinib 0-9 phosphatase and tensin homolog Homo sapiens 46-50 25776476-3 2015 Herein, we sought to evaluate the cytotoxic activity of sunitinib and everolimus against renal cancer cells Caki-1 and moreover to assess their impact on the expression levels of three BCL2 family members and three apoptosis-related microRNA clusters upon incubation with the drugs or following recovery from treatment. Sunitinib 56-65 BCL2 apoptosis regulator Homo sapiens 185-189 25776476-7 2015 In parallel, significant modulations were observed in the expression levels of miR-145, miR-15a, and miR-16 in case of sunitinib, whereas BCL2, BAX, miR-145 and miR-15a expression was strongly affected by everolimus. Sunitinib 119-128 microRNA 145 Homo sapiens 79-86 25776476-7 2015 In parallel, significant modulations were observed in the expression levels of miR-145, miR-15a, and miR-16 in case of sunitinib, whereas BCL2, BAX, miR-145 and miR-15a expression was strongly affected by everolimus. Sunitinib 119-128 microRNA 15a Homo sapiens 88-95 25260394-6 2015 The antitumor activity of hispidulin and its enhancement of the antitumor activity of sunitinib correlated with the suppression of pStat3 signaling and the consequent downregulation of Bcl-2 and survivin. Sunitinib 86-95 BCL2 apoptosis regulator Homo sapiens 185-190 25776476-7 2015 In parallel, significant modulations were observed in the expression levels of miR-145, miR-15a, and miR-16 in case of sunitinib, whereas BCL2, BAX, miR-145 and miR-15a expression was strongly affected by everolimus. Sunitinib 119-128 glycerophosphodiester phosphodiesterase 1 Homo sapiens 101-107 25776476-7 2015 In parallel, significant modulations were observed in the expression levels of miR-145, miR-15a, and miR-16 in case of sunitinib, whereas BCL2, BAX, miR-145 and miR-15a expression was strongly affected by everolimus. Sunitinib 119-128 microRNA 15a Homo sapiens 161-168 25776476-8 2015 Overall, our data support the notion that sunitinib and everolimus are able to directly induce cell death in renal cancer cells and simultaneously affect the expression levels of their apoptosis-related microRNAs and BCL2 family members upon this process. Sunitinib 42-51 BCL2 apoptosis regulator Homo sapiens 217-221 25663899-6 2015 Functionally, the role of ABCG2 in the resistance to sunitinib was confirmed by the use of the ABCG2 inhibitors fumitremorgin C and diethylstilbestrol, which blocked cell resistance. Sunitinib 53-62 ATP binding cassette subfamily G member 2 (Junior blood group) Mus musculus 26-31 25637219-0 2015 AXL as a modulator of sunitinib response in glioblastoma cell lines. Sunitinib 22-31 AXL receptor tyrosine kinase Homo sapiens 0-3 25637219-3 2015 We recently identified the RTK AXL as a putative target for the pan-RTK inhibitors cediranib and sunitinib, which are under clinical trials for glioblastoma patients. Sunitinib 97-106 ret proto-oncogene Homo sapiens 27-30 25637219-3 2015 We recently identified the RTK AXL as a putative target for the pan-RTK inhibitors cediranib and sunitinib, which are under clinical trials for glioblastoma patients. Sunitinib 97-106 AXL receptor tyrosine kinase Homo sapiens 31-34 25637219-3 2015 We recently identified the RTK AXL as a putative target for the pan-RTK inhibitors cediranib and sunitinib, which are under clinical trials for glioblastoma patients. Sunitinib 97-106 ret proto-oncogene Homo sapiens 68-71 25637219-4 2015 Here, we provide evidence that AXL activity can modulate sunitinib response in glioblastoma cell lines. Sunitinib 57-66 AXL receptor tyrosine kinase Homo sapiens 31-34 25637219-5 2015 We found that AXL knockdown conferred lower sensitivity to sunitinib by rescuing migratory defects and inhibiting apoptosis in cells expressing high AXL basal levels. Sunitinib 59-68 AXL receptor tyrosine kinase Homo sapiens 14-17 25637219-5 2015 We found that AXL knockdown conferred lower sensitivity to sunitinib by rescuing migratory defects and inhibiting apoptosis in cells expressing high AXL basal levels. Sunitinib 59-68 AXL receptor tyrosine kinase Homo sapiens 149-152 25637219-6 2015 Accordingly, overactivation of AXL by its ligand GAS6 rendered AXL positive glioblastoma cells more sensitive to sunitinib. Sunitinib 113-122 AXL receptor tyrosine kinase Homo sapiens 31-34 25637219-6 2015 Accordingly, overactivation of AXL by its ligand GAS6 rendered AXL positive glioblastoma cells more sensitive to sunitinib. Sunitinib 113-122 growth arrest specific 6 Homo sapiens 49-53 25637219-6 2015 Accordingly, overactivation of AXL by its ligand GAS6 rendered AXL positive glioblastoma cells more sensitive to sunitinib. Sunitinib 113-122 AXL receptor tyrosine kinase Homo sapiens 63-66 25637219-8 2015 The combination of sunitinib with a specific AKT inhibitor reverted the resistance of AXL-silenced cells to sunitinib. Sunitinib 19-28 AKT serine/threonine kinase 1 Homo sapiens 45-48 25637219-8 2015 The combination of sunitinib with a specific AKT inhibitor reverted the resistance of AXL-silenced cells to sunitinib. Sunitinib 19-28 AXL receptor tyrosine kinase Homo sapiens 86-89 25637219-8 2015 The combination of sunitinib with a specific AKT inhibitor reverted the resistance of AXL-silenced cells to sunitinib. Sunitinib 108-117 AKT serine/threonine kinase 1 Homo sapiens 45-48 25637219-8 2015 The combination of sunitinib with a specific AKT inhibitor reverted the resistance of AXL-silenced cells to sunitinib. Sunitinib 108-117 AXL receptor tyrosine kinase Homo sapiens 86-89 25637219-9 2015 Together, our results suggest that sunitinib inhibits AXL and AXL activation status modulates therapy response of glioblastoma cells to sunitinib. Sunitinib 35-44 AXL receptor tyrosine kinase Homo sapiens 54-57 25637219-9 2015 Together, our results suggest that sunitinib inhibits AXL and AXL activation status modulates therapy response of glioblastoma cells to sunitinib. Sunitinib 35-44 AXL receptor tyrosine kinase Homo sapiens 62-65 25637219-9 2015 Together, our results suggest that sunitinib inhibits AXL and AXL activation status modulates therapy response of glioblastoma cells to sunitinib. Sunitinib 136-145 AXL receptor tyrosine kinase Homo sapiens 62-65 25637219-10 2015 Moreover, it indicates that combining sunitinib therapy with AKT pathway inhibitors could overcome sunitinib resistance. Sunitinib 99-108 AKT serine/threonine kinase 1 Homo sapiens 61-64 25837361-9 2015 The expressions of Fas ligand and PARP in T24 cells treated with sunitinib malate exhibited a concentration-dependent increase. Sunitinib 65-81 Fas ligand Homo sapiens 19-29 25837361-9 2015 The expressions of Fas ligand and PARP in T24 cells treated with sunitinib malate exhibited a concentration-dependent increase. Sunitinib 65-81 poly(ADP-ribose) polymerase 1 Homo sapiens 34-38 25663899-6 2015 Functionally, the role of ABCG2 in the resistance to sunitinib was confirmed by the use of the ABCG2 inhibitors fumitremorgin C and diethylstilbestrol, which blocked cell resistance. Sunitinib 53-62 ATP binding cassette subfamily G member 2 (Junior blood group) Mus musculus 95-100 25675297-0 2015 p21-activated kinase 1 determines stem-like phenotype and sunitinib resistance via NF-kappaB/IL-6 activation in renal cell carcinoma. Sunitinib 58-67 nuclear factor kappa B subunit 1 Homo sapiens 83-92 25460504-0 2015 Sunitinib prevents cachexia and prolongs survival of mice bearing renal cancer by restraining STAT3 and MuRF-1 activation in muscle. Sunitinib 0-9 signal transducer and activator of transcription 3 Mus musculus 94-99 25460504-0 2015 Sunitinib prevents cachexia and prolongs survival of mice bearing renal cancer by restraining STAT3 and MuRF-1 activation in muscle. Sunitinib 0-9 tripartite motif-containing 63 Mus musculus 104-110 25460504-9 2015 Among the mechanisms, we herein show that sunitinib is able to restrain the overactivation of STAT3 and MuRF-1 pathways, involved in enhanced muscle protein catabolism during cancer cachexia. Sunitinib 42-51 signal transducer and activator of transcription 3 Mus musculus 94-99 25460504-9 2015 Among the mechanisms, we herein show that sunitinib is able to restrain the overactivation of STAT3 and MuRF-1 pathways, involved in enhanced muscle protein catabolism during cancer cachexia. Sunitinib 42-51 tripartite motif-containing 63 Mus musculus 104-110 25880253-0 2015 Melan-A/MART-1 immunity in a EWS-ATF1 translocated clear cell sarcoma patient treated with sunitinib: a case report. Sunitinib 91-100 melan-A Homo sapiens 0-7 25880253-0 2015 Melan-A/MART-1 immunity in a EWS-ATF1 translocated clear cell sarcoma patient treated with sunitinib: a case report. Sunitinib 91-100 melan-A Homo sapiens 8-14 25880253-0 2015 Melan-A/MART-1 immunity in a EWS-ATF1 translocated clear cell sarcoma patient treated with sunitinib: a case report. Sunitinib 91-100 activating transcription factor 1 Homo sapiens 29-37 25880253-4 2015 In the case we report here, treatment with sunitinib induced a long-lasting clinical response that was associated with an immune activation directed against Melan-A/MART-1 antigen. Sunitinib 43-52 melan-A Homo sapiens 157-164 25880253-4 2015 In the case we report here, treatment with sunitinib induced a long-lasting clinical response that was associated with an immune activation directed against Melan-A/MART-1 antigen. Sunitinib 43-52 melan-A Homo sapiens 165-171 25675297-7 2015 Furthermore, nuclear factor-kappaB (NF-kappaB)/interleukin-6 (IL-6) activation was found to be responsible for PAK1-mediated stem-like phenotype following sunitinib treatment. Sunitinib 155-164 interleukin 6 Homo sapiens 62-66 25675297-7 2015 Furthermore, nuclear factor-kappaB (NF-kappaB)/interleukin-6 (IL-6) activation was found to be responsible for PAK1-mediated stem-like phenotype following sunitinib treatment. Sunitinib 155-164 p21 (RAC1) activated kinase 1 Homo sapiens 111-115 25675297-8 2015 Both IL-6 neutralizing antibody and IPA3 administration enhanced tumor growth inhibition effect of sunitinib treatment on RCC cells in vitro and in vivo. Sunitinib 99-108 interleukin 6 Homo sapiens 5-9 25675297-9 2015 Our results unraveled that oncogenic activation of PAK1 defines an important mechanism for maintaining stem-like phenotype and sunitinib resistance through NF-kappaB/IL-6 activation in RCC, lending PAK1-mediated NF-kappaB/IL-6 activation considerable appeal as novel pharmacological therapeutic targets against sunitinib resistance. Sunitinib 127-136 p21 (RAC1) activated kinase 1 Homo sapiens 51-55 25675297-9 2015 Our results unraveled that oncogenic activation of PAK1 defines an important mechanism for maintaining stem-like phenotype and sunitinib resistance through NF-kappaB/IL-6 activation in RCC, lending PAK1-mediated NF-kappaB/IL-6 activation considerable appeal as novel pharmacological therapeutic targets against sunitinib resistance. Sunitinib 127-136 nuclear factor kappa B subunit 1 Homo sapiens 156-165 25675297-9 2015 Our results unraveled that oncogenic activation of PAK1 defines an important mechanism for maintaining stem-like phenotype and sunitinib resistance through NF-kappaB/IL-6 activation in RCC, lending PAK1-mediated NF-kappaB/IL-6 activation considerable appeal as novel pharmacological therapeutic targets against sunitinib resistance. Sunitinib 127-136 interleukin 6 Homo sapiens 166-170 25675297-9 2015 Our results unraveled that oncogenic activation of PAK1 defines an important mechanism for maintaining stem-like phenotype and sunitinib resistance through NF-kappaB/IL-6 activation in RCC, lending PAK1-mediated NF-kappaB/IL-6 activation considerable appeal as novel pharmacological therapeutic targets against sunitinib resistance. Sunitinib 127-136 nuclear factor kappa B subunit 1 Homo sapiens 212-221 25675297-0 2015 p21-activated kinase 1 determines stem-like phenotype and sunitinib resistance via NF-kappaB/IL-6 activation in renal cell carcinoma. Sunitinib 58-67 interleukin 6 Homo sapiens 93-97 25675297-9 2015 Our results unraveled that oncogenic activation of PAK1 defines an important mechanism for maintaining stem-like phenotype and sunitinib resistance through NF-kappaB/IL-6 activation in RCC, lending PAK1-mediated NF-kappaB/IL-6 activation considerable appeal as novel pharmacological therapeutic targets against sunitinib resistance. Sunitinib 127-136 interleukin 6 Homo sapiens 222-226 25675297-9 2015 Our results unraveled that oncogenic activation of PAK1 defines an important mechanism for maintaining stem-like phenotype and sunitinib resistance through NF-kappaB/IL-6 activation in RCC, lending PAK1-mediated NF-kappaB/IL-6 activation considerable appeal as novel pharmacological therapeutic targets against sunitinib resistance. Sunitinib 311-320 p21 (RAC1) activated kinase 1 Homo sapiens 51-55 25675297-6 2015 Stem-like phenotype due to sunitinib administration via increased PAK1 kinase activation could be ameliorated by PAK1 shRNA, PAK1 mutant K299R and IPA3. Sunitinib 27-36 p21 (RAC1) activated kinase 1 Homo sapiens 66-70 25675297-9 2015 Our results unraveled that oncogenic activation of PAK1 defines an important mechanism for maintaining stem-like phenotype and sunitinib resistance through NF-kappaB/IL-6 activation in RCC, lending PAK1-mediated NF-kappaB/IL-6 activation considerable appeal as novel pharmacological therapeutic targets against sunitinib resistance. Sunitinib 311-320 nuclear factor kappa B subunit 1 Homo sapiens 156-165 25675297-9 2015 Our results unraveled that oncogenic activation of PAK1 defines an important mechanism for maintaining stem-like phenotype and sunitinib resistance through NF-kappaB/IL-6 activation in RCC, lending PAK1-mediated NF-kappaB/IL-6 activation considerable appeal as novel pharmacological therapeutic targets against sunitinib resistance. Sunitinib 311-320 interleukin 6 Homo sapiens 166-170 25675297-9 2015 Our results unraveled that oncogenic activation of PAK1 defines an important mechanism for maintaining stem-like phenotype and sunitinib resistance through NF-kappaB/IL-6 activation in RCC, lending PAK1-mediated NF-kappaB/IL-6 activation considerable appeal as novel pharmacological therapeutic targets against sunitinib resistance. Sunitinib 311-320 p21 (RAC1) activated kinase 1 Homo sapiens 198-202 25675297-6 2015 Stem-like phenotype due to sunitinib administration via increased PAK1 kinase activation could be ameliorated by PAK1 shRNA, PAK1 mutant K299R and IPA3. Sunitinib 27-36 p21 (RAC1) activated kinase 1 Homo sapiens 113-117 25675297-9 2015 Our results unraveled that oncogenic activation of PAK1 defines an important mechanism for maintaining stem-like phenotype and sunitinib resistance through NF-kappaB/IL-6 activation in RCC, lending PAK1-mediated NF-kappaB/IL-6 activation considerable appeal as novel pharmacological therapeutic targets against sunitinib resistance. Sunitinib 311-320 nuclear factor kappa B subunit 1 Homo sapiens 212-221 25675297-9 2015 Our results unraveled that oncogenic activation of PAK1 defines an important mechanism for maintaining stem-like phenotype and sunitinib resistance through NF-kappaB/IL-6 activation in RCC, lending PAK1-mediated NF-kappaB/IL-6 activation considerable appeal as novel pharmacological therapeutic targets against sunitinib resistance. Sunitinib 311-320 interleukin 6 Homo sapiens 222-226 25675297-6 2015 Stem-like phenotype due to sunitinib administration via increased PAK1 kinase activation could be ameliorated by PAK1 shRNA, PAK1 mutant K299R and IPA3. Sunitinib 27-36 p21 (RAC1) activated kinase 1 Homo sapiens 113-117 25675301-1 2015 The angiogenesis inhibitor sunitinib is a tyrosine kinase inhibitor that acts mainly on the VEGF and PDGF pathways. Sunitinib 27-36 vascular endothelial growth factor A Mus musculus 92-96 25675297-7 2015 Furthermore, nuclear factor-kappaB (NF-kappaB)/interleukin-6 (IL-6) activation was found to be responsible for PAK1-mediated stem-like phenotype following sunitinib treatment. Sunitinib 155-164 nuclear factor kappa B subunit 1 Homo sapiens 13-34 25675297-7 2015 Furthermore, nuclear factor-kappaB (NF-kappaB)/interleukin-6 (IL-6) activation was found to be responsible for PAK1-mediated stem-like phenotype following sunitinib treatment. Sunitinib 155-164 nuclear factor kappa B subunit 1 Homo sapiens 36-46 25675297-7 2015 Furthermore, nuclear factor-kappaB (NF-kappaB)/interleukin-6 (IL-6) activation was found to be responsible for PAK1-mediated stem-like phenotype following sunitinib treatment. Sunitinib 155-164 interleukin 6 Homo sapiens 47-60 25363319-7 2015 Tumour growth and intratumour VEGF receptor-2 expressions were significantly reduced in sunitinib-treated mice, while the expression of the other targeted receptors, PDGF receptor -alpha or -beta and fibroblast growth factor receptor-1, remained unaffected. Sunitinib 88-97 kinase insert domain protein receptor Mus musculus 30-45 25363319-9 2015 There were intratumour areas where the signal intensity of sunitinib correlated with expression of VEGF receptor-2. Sunitinib 59-68 kinase insert domain protein receptor Mus musculus 99-114 25344452-9 2015 The LBM (p < 0.0001) and a polymorphism in the ABCG2 transporter (421C>A) (p = 0.014) were two independent parameters accounting for the variability of composite (sunitinib + SU12662) exposure. Sunitinib 169-178 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 50-55 25458015-10 2015 We determined the underlying pathways by which sunitinib operates as a toxin on gliomas and found vascular endothelial growth factor receptor 2 (VEGFR2, KDR/Flk1) as the main target to execute gliomatoxicity. Sunitinib 47-56 kinase insert domain receptor Homo sapiens 98-143 25458015-10 2015 We determined the underlying pathways by which sunitinib operates as a toxin on gliomas and found vascular endothelial growth factor receptor 2 (VEGFR2, KDR/Flk1) as the main target to execute gliomatoxicity. Sunitinib 47-56 kinase insert domain receptor Homo sapiens 153-156 25458015-10 2015 We determined the underlying pathways by which sunitinib operates as a toxin on gliomas and found vascular endothelial growth factor receptor 2 (VEGFR2, KDR/Flk1) as the main target to execute gliomatoxicity. Sunitinib 47-56 kinase insert domain receptor Homo sapiens 157-161 25458015-11 2015 The apoptosis-inducing effect of sunitinib can be mimicked by inhibition of VEGFR2. Sunitinib 33-42 kinase insert domain receptor Homo sapiens 76-82 24403097-5 2015 RESULTS: Sunitinib interacts with CYP3A4 inducers or inhibitors and with P-glycoprotein and ABCG2 substrates. Sunitinib 9-18 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 92-97 25444514-10 2015 It is concluded that an mTOR-inhibitor for second-line therapy could be the strategy of choice after first-line sunitinib failure. Sunitinib 112-121 mechanistic target of rapamycin kinase Homo sapiens 24-28 25422908-1 2015 BACKGROUND: Sunitinib (VEGFR/PDGFR inhibitor) and everolimus (mTOR inhibitor) are both approved for advanced renal cell carcinoma (RCC) as first-line and second-line therapy, respectively. Sunitinib 12-21 kinase insert domain receptor Homo sapiens 23-28 25519701-12 2015 Furthermore, specific EZH2 inhibition resulted in increased in vitro antitumor effect of sunitinib. Sunitinib 89-98 enhancer of zeste 2 polycomb repressive complex 2 subunit Homo sapiens 22-26 25437118-5 2015 We found that chronic restraint stress markedly weakened the efficacy of sunitinib, primarily through promoting the expression of vascular endothelial growth factor (VEGF) and interleukin-8 (IL-8) to stimulate tumor angiogenesis in vivo. Sunitinib 73-82 vascular endothelial growth factor A Mus musculus 130-164 25437118-5 2015 We found that chronic restraint stress markedly weakened the efficacy of sunitinib, primarily through promoting the expression of vascular endothelial growth factor (VEGF) and interleukin-8 (IL-8) to stimulate tumor angiogenesis in vivo. Sunitinib 73-82 vascular endothelial growth factor A Mus musculus 166-170 25437118-5 2015 We found that chronic restraint stress markedly weakened the efficacy of sunitinib, primarily through promoting the expression of vascular endothelial growth factor (VEGF) and interleukin-8 (IL-8) to stimulate tumor angiogenesis in vivo. Sunitinib 73-82 chemokine (C-X-C motif) ligand 15 Mus musculus 176-189 25437118-5 2015 We found that chronic restraint stress markedly weakened the efficacy of sunitinib, primarily through promoting the expression of vascular endothelial growth factor (VEGF) and interleukin-8 (IL-8) to stimulate tumor angiogenesis in vivo. Sunitinib 73-82 chemokine (C-X-C motif) ligand 15 Mus musculus 191-195 25437118-7 2015 In vitro, norepinephrine up-regulated expression of VEGF and IL-8 in sunitinib-treated cancer cells mainly through the beta-adrenoceptor-cAMP-PKA signaling pathway. Sunitinib 69-78 vascular endothelial growth factor A Mus musculus 52-56 25437118-7 2015 In vitro, norepinephrine up-regulated expression of VEGF and IL-8 in sunitinib-treated cancer cells mainly through the beta-adrenoceptor-cAMP-PKA signaling pathway. Sunitinib 69-78 chemokine (C-X-C motif) ligand 15 Mus musculus 61-65 25422908-1 2015 BACKGROUND: Sunitinib (VEGFR/PDGFR inhibitor) and everolimus (mTOR inhibitor) are both approved for advanced renal cell carcinoma (RCC) as first-line and second-line therapy, respectively. Sunitinib 12-21 platelet derived growth factor receptor beta Homo sapiens 29-34 25455500-6 2015 We hypothesized that inhibition of multidrug resistant transporters by elacridar (dual inhibitor of P-glycoprotein and ABCG 2) might overcome sunitinib resistance in experimental renal cell carcinoma. Sunitinib 142-151 ATP binding cassette subfamily B member 1 Homo sapiens 100-114 25455500-6 2015 We hypothesized that inhibition of multidrug resistant transporters by elacridar (dual inhibitor of P-glycoprotein and ABCG 2) might overcome sunitinib resistance in experimental renal cell carcinoma. Sunitinib 142-151 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 119-125 25455500-11 2015 ABCG2 function was inhibited by sunitinib alone or combination with elacridar but not elacridar alone. Sunitinib 32-41 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 0-5 25455500-12 2015 These findings suggest that sunitinib resistance involves multidrug resistance transporters, and in combination with elacridar, can be reversed in renal carcinoma cells by P-glycoprotein inhibition. Sunitinib 28-37 ATP binding cassette subfamily B member 1 Homo sapiens 172-186 26064968-0 2015 miR-155 and miR-484 Are Associated with Time to Progression in Metastatic Renal Cell Carcinoma Treated with Sunitinib. Sunitinib 108-117 microRNA 155 Homo sapiens 0-7 26312386-6 2015 At these concentrations, sunitinib accumulated in lysosomes, which downregulated the activity of the lysosomal protease CTSB (cathepsin B) and led to incomplete autophagic flux. Sunitinib 25-34 cathepsin B Homo sapiens 120-124 26312386-6 2015 At these concentrations, sunitinib accumulated in lysosomes, which downregulated the activity of the lysosomal protease CTSB (cathepsin B) and led to incomplete autophagic flux. Sunitinib 25-34 cathepsin B Homo sapiens 126-137 26312386-8 2015 Sunitinib stimulated the expression of ABCB1 (ATP-binding cassette, sub-family B [MDR/TAP], member 1), which participates in the accumulation of the drug in autolysosomes and favor its cellular efflux. Sunitinib 0-9 ATP binding cassette subfamily B member 1 Homo sapiens 39-44 26312386-8 2015 Sunitinib stimulated the expression of ABCB1 (ATP-binding cassette, sub-family B [MDR/TAP], member 1), which participates in the accumulation of the drug in autolysosomes and favor its cellular efflux. Sunitinib 0-9 ATP binding cassette subfamily B member 1 Homo sapiens 46-100 25446042-2 2015 Sunitinib, a tyrosine kinase inhibitor of the VEGF receptor, has become the mainstay of treatment for these patients. Sunitinib 0-9 vascular endothelial growth factor A Homo sapiens 46-50 25993161-7 2015 For recurrent and aggressive tumors, inhibitors of the vascular endothelial growth factor (VEGF) pathway, such as vatalinib, bevacizumab, and sunitinib, showed signs of activity in small, uncontrolled studies, and prospective clinical studies will test the efficacy of the tetrahydroisoquinoline trabectedin and of SMO and AKT1 inhibitors. Sunitinib 142-151 vascular endothelial growth factor A Homo sapiens 55-89 25993161-7 2015 For recurrent and aggressive tumors, inhibitors of the vascular endothelial growth factor (VEGF) pathway, such as vatalinib, bevacizumab, and sunitinib, showed signs of activity in small, uncontrolled studies, and prospective clinical studies will test the efficacy of the tetrahydroisoquinoline trabectedin and of SMO and AKT1 inhibitors. Sunitinib 142-151 vascular endothelial growth factor A Homo sapiens 91-95 25993161-7 2015 For recurrent and aggressive tumors, inhibitors of the vascular endothelial growth factor (VEGF) pathway, such as vatalinib, bevacizumab, and sunitinib, showed signs of activity in small, uncontrolled studies, and prospective clinical studies will test the efficacy of the tetrahydroisoquinoline trabectedin and of SMO and AKT1 inhibitors. Sunitinib 142-151 smoothened, frizzled class receptor Homo sapiens 315-318 25993161-7 2015 For recurrent and aggressive tumors, inhibitors of the vascular endothelial growth factor (VEGF) pathway, such as vatalinib, bevacizumab, and sunitinib, showed signs of activity in small, uncontrolled studies, and prospective clinical studies will test the efficacy of the tetrahydroisoquinoline trabectedin and of SMO and AKT1 inhibitors. Sunitinib 142-151 AKT serine/threonine kinase 1 Homo sapiens 323-327 26320436-8 2015 Six month disease control rate (complete response, partial response and stable disease) in response to sunitinib was significantly higher in patients with normal (<=0.5 mg/dl) than elevated baseline CRP (p<0.001). Sunitinib 103-112 C-reactive protein Homo sapiens 202-205 26320436-9 2015 CONCLUSIONS: CRP is a significant independent predictor of PFS for Japanese patients with mCCRCC treated with first-line sunitinib. Sunitinib 121-130 C-reactive protein Homo sapiens 13-16 26320436-10 2015 Pretreatment CRP concentration may be a useful biomarker predicting response to sunitinib treatment. Sunitinib 80-89 C-reactive protein Homo sapiens 13-16 26064968-0 2015 miR-155 and miR-484 Are Associated with Time to Progression in Metastatic Renal Cell Carcinoma Treated with Sunitinib. Sunitinib 108-117 microRNA 484 Homo sapiens 12-19 26064968-12 2015 miR-155 and miR-484 are potentially connected with sunitinib resistance and failure of the therapy. Sunitinib 51-60 microRNA 155 Homo sapiens 0-7 26064968-12 2015 miR-155 and miR-484 are potentially connected with sunitinib resistance and failure of the therapy. Sunitinib 51-60 microRNA 484 Homo sapiens 12-19 26788996-5 2015 Sunitinib inhibited PDGF-stimulated proliferation, migration, phosphorylation of MAPK and PI3K/Akt proteins and changes in the expression of cell-cycle regulatory proteins in vascular smooth-muscle cells as well as VEGF-stimulated endothelial cell proliferation in vitro. Sunitinib 0-9 AKT serine/threonine kinase 1 Homo sapiens 95-98 25376776-7 2015 In the presence of VEGF stimulation, the ROCK inhibitor suppressed stretch-induced sprout alignment but did not affect stretch-induced sprout density; in contrast, the receptor tyrosine kinase (RTK) inhibitor sunitinib had no effect on stretch-induced alignment but trended toward suppressed stretch-induced sprout density. Sunitinib 209-218 ret proto-oncogene Homo sapiens 194-197 24865476-13 2015 Sunitinib also blocked LRP1 phosphorylation in response to PDGF-BB and induced phosphorylation of ERK, but this latter event was not affected by LRP1 knockdown. Sunitinib 0-9 LDL receptor related protein 1 Rattus norvegicus 23-27 24865476-13 2015 Sunitinib also blocked LRP1 phosphorylation in response to PDGF-BB and induced phosphorylation of ERK, but this latter event was not affected by LRP1 knockdown. Sunitinib 0-9 Eph receptor B1 Rattus norvegicus 98-101 25361735-6 2015 Inhibition of VEGF signaling in B16 melanoma tumor-bearing mice by sunitinib treatment resulted in up-regulation of CXCL10 and CXCL11 in tumor vessels, accompanied by up to 18-fold increased infiltration of CD3(+) T-lymphocytes in B16 tumors. Sunitinib 67-76 vascular endothelial growth factor A Mus musculus 14-18 25361735-6 2015 Inhibition of VEGF signaling in B16 melanoma tumor-bearing mice by sunitinib treatment resulted in up-regulation of CXCL10 and CXCL11 in tumor vessels, accompanied by up to 18-fold increased infiltration of CD3(+) T-lymphocytes in B16 tumors. Sunitinib 67-76 chemokine (C-X-C motif) ligand 10 Mus musculus 116-122 25361735-6 2015 Inhibition of VEGF signaling in B16 melanoma tumor-bearing mice by sunitinib treatment resulted in up-regulation of CXCL10 and CXCL11 in tumor vessels, accompanied by up to 18-fold increased infiltration of CD3(+) T-lymphocytes in B16 tumors. Sunitinib 67-76 chemokine (C-X-C motif) ligand 11 Mus musculus 127-133 26788996-5 2015 Sunitinib inhibited PDGF-stimulated proliferation, migration, phosphorylation of MAPK and PI3K/Akt proteins and changes in the expression of cell-cycle regulatory proteins in vascular smooth-muscle cells as well as VEGF-stimulated endothelial cell proliferation in vitro. Sunitinib 0-9 vascular endothelial growth factor A Homo sapiens 215-219 25381264-6 2015 Treatment with single-agent crizotinib reduced tumor vascularization but failed to inhibit tumor growth in either model, despite also a significant increase of c-met expression and phosphorylation in the sunitinib-resistant tumors. Sunitinib 204-213 MET proto-oncogene, receptor tyrosine kinase Homo sapiens 160-165 25100872-2 2015 Sunitinib malate (SU011248) is a small-molecule tyrosine kinase inhibitor that targets vascular endothelial growth factor receptor (VEGFR) and platelet-derived growth factor receptor, abundant in meningiomas. Sunitinib 0-16 kinase insert domain receptor Homo sapiens 87-130 25519698-4 2015 Axitinib, pazopanib, and sunitinib inhibited hENT1 at low micromolar concentrations. Sunitinib 25-34 solute carrier family 29 member 1 (Augustine blood group) Homo sapiens 45-50 25519698-6 2015 Pazopanib > axitinib >= sunitinib inhibited hENT1 with IC50 values of 2, 7, and 29 mumol/L, respectively, leading to reduced intracellular gemcitabine and FLT accumulation. Sunitinib 30-39 solute carrier family 29 member 1 (Augustine blood group) Homo sapiens 50-55 25100872-2 2015 Sunitinib malate (SU011248) is a small-molecule tyrosine kinase inhibitor that targets vascular endothelial growth factor receptor (VEGFR) and platelet-derived growth factor receptor, abundant in meningiomas. Sunitinib 0-16 kinase insert domain receptor Homo sapiens 132-137 25100872-2 2015 Sunitinib malate (SU011248) is a small-molecule tyrosine kinase inhibitor that targets vascular endothelial growth factor receptor (VEGFR) and platelet-derived growth factor receptor, abundant in meningiomas. Sunitinib 18-26 kinase insert domain receptor Homo sapiens 87-130 25100872-2 2015 Sunitinib malate (SU011248) is a small-molecule tyrosine kinase inhibitor that targets vascular endothelial growth factor receptor (VEGFR) and platelet-derived growth factor receptor, abundant in meningiomas. Sunitinib 18-26 kinase insert domain receptor Homo sapiens 132-137 24710685-5 2015 VEGFR-2 expression was associated significantly with initial and best overall responses to sunitinib, along with Karnofsky performance status and Memorial Sloan-Kettering Cancer Center prognostic risk group. Sunitinib 91-100 kinase insert domain receptor Homo sapiens 0-7 25575050-0 2015 [Inhibition of sunitinib on the expressions of PD-L1 and PD-L2 of mouse bone marrow-derived dendritic cells]. Sunitinib 15-24 CD274 antigen Mus musculus 47-52 25575050-0 2015 [Inhibition of sunitinib on the expressions of PD-L1 and PD-L2 of mouse bone marrow-derived dendritic cells]. Sunitinib 15-24 programmed cell death 1 ligand 2 Mus musculus 57-62 25575050-1 2015 OBJECTIVE: To observe the changes of programmed death-1 ligand 1 (PD-L1) and PD-L2 expressions on mouse bone marrow-derived dendritic cells (DCs) stimulated by sunitinib. Sunitinib 160-169 CD274 antigen Mus musculus 37-64 24710685-9 2015 CONCLUSION: VEGFR-2 expression might be a useful biomarker for predicting the response to sunitinib by patients with metastatic RCC. Sunitinib 90-99 kinase insert domain receptor Homo sapiens 12-19 25575050-1 2015 OBJECTIVE: To observe the changes of programmed death-1 ligand 1 (PD-L1) and PD-L2 expressions on mouse bone marrow-derived dendritic cells (DCs) stimulated by sunitinib. Sunitinib 160-169 CD274 antigen Mus musculus 66-71 25575050-1 2015 OBJECTIVE: To observe the changes of programmed death-1 ligand 1 (PD-L1) and PD-L2 expressions on mouse bone marrow-derived dendritic cells (DCs) stimulated by sunitinib. Sunitinib 160-169 programmed cell death 1 ligand 2 Mus musculus 77-82 25621299-9 2014 The lysosomal capacity reflected by LAMP-1 and -2 expression was higher in HT-29SUN compared to HT-29PAR tumors indicating an increased sequestration of sunitinib in lysosomes of resistant tumors. Sunitinib 153-162 lysosomal associated membrane protein 1 Homo sapiens 36-49 25575050-4 2015 RESULTS: Compared with the control group, the expression of PD-L1 on mature DCs (mDCs) and all DCs [including mature DCs and immature DCs (imDCs)] was significantly down-regulated in sunitinib treatment groups. Sunitinib 183-192 CD274 antigen Mus musculus 60-65 25575050-5 2015 The PD-L1 expression percentages of imDCs, mDCs and DCs were significantly reduced in sunitinib treatment groups; the percentage of mDCs expressing PD-L2 also dropped in all treatment groups, and the percentage of DCs expressing PD-L2 decreased in 100 and 300 ng/mL sunitinib treatment groups. Sunitinib 86-95 CD274 antigen Mus musculus 4-9 25575050-6 2015 CONCLUSION: Sunitinib can significantly reduce the expressions of PD-L1 and PD-L2 on mouse DCs. Sunitinib 12-21 CD274 antigen Mus musculus 66-71 25575050-6 2015 CONCLUSION: Sunitinib can significantly reduce the expressions of PD-L1 and PD-L2 on mouse DCs. Sunitinib 12-21 programmed cell death 1 ligand 2 Mus musculus 76-81 25540214-3 2014 When started on sunitinib he experienced improvement in his diabetes control with reduction in his insulin requirements, which later worsened when sunitinib was reduced or stopped. Sunitinib 16-25 insulin Homo sapiens 99-106 25515134-0 2014 Sunitinib-induced severe toxicities in a Japanese patient with the ABCG2 421 AA genotype. Sunitinib 0-9 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 67-72 25515134-1 2014 BACKGROUND: Sunitinib is a multi-targeted receptor tyrosine kinase inhibitor that acts against receptors for vascular endothelial growth factor and platelet-derived growth factor. Sunitinib 12-21 vascular endothelial growth factor A Homo sapiens 109-143 25515134-10 2014 The patient"s genotype of ABCG2 (ATP-binding cassette, sub-family G (WHITE), member 2) 421C > A was homozygous for the variant allele (AA), which was reported to be associated with high exposure to sunitinib. Sunitinib 201-210 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 26-31 25515134-11 2014 Therefore, we speculated that the extremely high plasma concentrations of sunitinib and SU12662 caused by the ABCG2 421 AA genotype might have resulted in severe toxicities to the patient. Sunitinib 74-83 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 110-115 25461627-8 2014 CONCLUSION: The orally-available XPO1 inhibitor, KPT-330, represents a novel target for RCC whose in vivo efficacy approaches that of sunitinib. Sunitinib 134-143 exportin 1 Homo sapiens 33-37 25353068-5 2014 Our results demonstrated that a significantly higher amount of sunitinib could be delivered to the U87MG tumor by targeting VEGFR when compared with the non-targeted counterparts. Sunitinib 63-72 kinase insert domain receptor Homo sapiens 124-129 25117211-7 2014 KEY RESULTS: Among 12 different compounds, sunitinib inhibited CDK5 with an IC50 of 4.2 muM. Sunitinib 43-52 cyclin dependent kinase 5 Homo sapiens 63-67 25117211-8 2014 In silico analysis revealed that, similarly to roscovitine, sunitinib fitted 6 of 10 features of the CDK5 pharmacophore. Sunitinib 60-69 cyclin dependent kinase 5 Homo sapiens 101-105 25117211-9 2014 In a cell-based model, sunitinib reduced CDK5 phosphorylation (pCDK5), calpain-dependent p35/p25 conversion and protected neuronal cells from the toxic effects of gp120. Sunitinib 23-32 cyclin dependent kinase 5 Homo sapiens 41-45 25117211-9 2014 In a cell-based model, sunitinib reduced CDK5 phosphorylation (pCDK5), calpain-dependent p35/p25 conversion and protected neuronal cells from the toxic effects of gp120. Sunitinib 23-32 cyclin dependent kinase 5 regulatory subunit 1 Homo sapiens 89-92 25117211-9 2014 In a cell-based model, sunitinib reduced CDK5 phosphorylation (pCDK5), calpain-dependent p35/p25 conversion and protected neuronal cells from the toxic effects of gp120. Sunitinib 23-32 cyclin dependent kinase 5 regulatory subunit 1 Homo sapiens 93-96 25117211-9 2014 In a cell-based model, sunitinib reduced CDK5 phosphorylation (pCDK5), calpain-dependent p35/p25 conversion and protected neuronal cells from the toxic effects of gp120. Sunitinib 23-32 inter-alpha-trypsin inhibitor heavy chain 4 Homo sapiens 163-168 25117211-10 2014 In glial fibrillary acidic protein-gp120 transgenic (tg) mice, sunitinib reduced levels of pCDK5, p35/p25 and phosphorylated tau protein, along with amelioration of the neurodegenerative pathology. Sunitinib 63-72 inter-alpha-trypsin inhibitor heavy chain 4 Homo sapiens 35-40 25117211-10 2014 In glial fibrillary acidic protein-gp120 transgenic (tg) mice, sunitinib reduced levels of pCDK5, p35/p25 and phosphorylated tau protein, along with amelioration of the neurodegenerative pathology. Sunitinib 63-72 cyclin-dependent kinase 5, regulatory subunit 1 (p35) Mus musculus 98-101 25117211-10 2014 In glial fibrillary acidic protein-gp120 transgenic (tg) mice, sunitinib reduced levels of pCDK5, p35/p25 and phosphorylated tau protein, along with amelioration of the neurodegenerative pathology. Sunitinib 63-72 cyclin-dependent kinase 5, regulatory subunit 1 (p35) Mus musculus 102-105 25151214-1 2014 Sunitinib malate (Sutent; Pfizer, Inc., New York, NY) is an oral multitargeted tyrosine kinase inhibitor that inhibits VEGF receptors (VEGFR-1, VEGFR-2, and VEGFR-3) among a family of kinase targets and have a central role in first-line treatment of metastatic renal cell carcinoma (mRCC). Sunitinib 0-16 fms related receptor tyrosine kinase 1 Homo sapiens 135-142 25151214-1 2014 Sunitinib malate (Sutent; Pfizer, Inc., New York, NY) is an oral multitargeted tyrosine kinase inhibitor that inhibits VEGF receptors (VEGFR-1, VEGFR-2, and VEGFR-3) among a family of kinase targets and have a central role in first-line treatment of metastatic renal cell carcinoma (mRCC). Sunitinib 0-16 kinase insert domain receptor Homo sapiens 144-151 25151214-1 2014 Sunitinib malate (Sutent; Pfizer, Inc., New York, NY) is an oral multitargeted tyrosine kinase inhibitor that inhibits VEGF receptors (VEGFR-1, VEGFR-2, and VEGFR-3) among a family of kinase targets and have a central role in first-line treatment of metastatic renal cell carcinoma (mRCC). Sunitinib 0-16 fms related receptor tyrosine kinase 4 Homo sapiens 157-164 25151214-1 2014 Sunitinib malate (Sutent; Pfizer, Inc., New York, NY) is an oral multitargeted tyrosine kinase inhibitor that inhibits VEGF receptors (VEGFR-1, VEGFR-2, and VEGFR-3) among a family of kinase targets and have a central role in first-line treatment of metastatic renal cell carcinoma (mRCC). Sunitinib 18-24 fms related receptor tyrosine kinase 1 Homo sapiens 135-142 25151214-1 2014 Sunitinib malate (Sutent; Pfizer, Inc., New York, NY) is an oral multitargeted tyrosine kinase inhibitor that inhibits VEGF receptors (VEGFR-1, VEGFR-2, and VEGFR-3) among a family of kinase targets and have a central role in first-line treatment of metastatic renal cell carcinoma (mRCC). Sunitinib 18-24 kinase insert domain receptor Homo sapiens 144-151 25151214-1 2014 Sunitinib malate (Sutent; Pfizer, Inc., New York, NY) is an oral multitargeted tyrosine kinase inhibitor that inhibits VEGF receptors (VEGFR-1, VEGFR-2, and VEGFR-3) among a family of kinase targets and have a central role in first-line treatment of metastatic renal cell carcinoma (mRCC). Sunitinib 18-24 fms related receptor tyrosine kinase 4 Homo sapiens 157-164 25299731-1 2014 A detailed atomistic description of the unbinding process of sorafenib and sunitinib, two known VEGFR2 inhibitors clinically used to treat renal cell carcinoma, was unraveled by using steered molecular dynamics (SMD) simulations. Sunitinib 75-84 kinase insert domain receptor Homo sapiens 96-102 24424564-1 2014 Sunitinib is a tyrosine kinase inhibitor with direct anti-tumor and anti-angiogenesis activity targeting VEGFR 1-2, PDGFR alpha-beta, c-kit, bFGF, (CSF-1), FLT3 and RET. Sunitinib 0-9 fms related receptor tyrosine kinase 1 Homo sapiens 105-112 24424564-1 2014 Sunitinib is a tyrosine kinase inhibitor with direct anti-tumor and anti-angiogenesis activity targeting VEGFR 1-2, PDGFR alpha-beta, c-kit, bFGF, (CSF-1), FLT3 and RET. Sunitinib 0-9 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 134-139 24424564-1 2014 Sunitinib is a tyrosine kinase inhibitor with direct anti-tumor and anti-angiogenesis activity targeting VEGFR 1-2, PDGFR alpha-beta, c-kit, bFGF, (CSF-1), FLT3 and RET. Sunitinib 0-9 fibroblast growth factor 2 Homo sapiens 141-145 24424564-1 2014 Sunitinib is a tyrosine kinase inhibitor with direct anti-tumor and anti-angiogenesis activity targeting VEGFR 1-2, PDGFR alpha-beta, c-kit, bFGF, (CSF-1), FLT3 and RET. Sunitinib 0-9 colony stimulating factor 1 Homo sapiens 148-153 24424564-1 2014 Sunitinib is a tyrosine kinase inhibitor with direct anti-tumor and anti-angiogenesis activity targeting VEGFR 1-2, PDGFR alpha-beta, c-kit, bFGF, (CSF-1), FLT3 and RET. Sunitinib 0-9 fms related receptor tyrosine kinase 3 Homo sapiens 156-160 24424564-1 2014 Sunitinib is a tyrosine kinase inhibitor with direct anti-tumor and anti-angiogenesis activity targeting VEGFR 1-2, PDGFR alpha-beta, c-kit, bFGF, (CSF-1), FLT3 and RET. Sunitinib 0-9 ret proto-oncogene Homo sapiens 165-168 25239608-2 2014 Imatinib, sunitinib, and regorafenib are approved therapies; however, efficacy is often limited by the acquisition of polyclonal secondary resistance mutations in KIT, with those located in the activation (A) loop (exons 17/18) being particularly problematic. Sunitinib 10-19 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 163-166 24802849-0 2014 Fas/Fas ligand mediates keratinocyte death in sunitinib-induced hand-foot skin reaction. Sunitinib 46-55 Fas ligand Homo sapiens 4-14 24821582-13 2014 Array CGH profiles revealed sunitinib was associated with significant CA9 region loss. Sunitinib 28-37 carbonic anhydrase 9 Homo sapiens 70-73 24821582-14 2014 RNAi CA9 silencing in two cell lines inhibited the antiproliferative effects of sunitinib. Sunitinib 80-89 carbonic anhydrase 9 Homo sapiens 5-8 25691928-4 2014 The inhibitory effects of CEP701, imatinib, dasatinib, PKC412 and sunitinib were studied on cell proliferation and FLT3 tyrosine phosphorylation. Sunitinib 66-75 fms related receptor tyrosine kinase 3 Homo sapiens 115-119 25691928-8 2014 FD-FLT3-ITD cells were three times more resistant to sunitinib than the FD-FLT3-WT cells. Sunitinib 53-62 fms related receptor tyrosine kinase 3 Homo sapiens 3-7 25275248-1 2014 OBJECTIVES: The therapeutic use of the vascular endothelial growth factor (VEGF) antagonist sunitinib is limited by sunitinib-induced hypertension. Sunitinib 92-101 vascular endothelial growth factor A Rattus norvegicus 39-73 25275248-1 2014 OBJECTIVES: The therapeutic use of the vascular endothelial growth factor (VEGF) antagonist sunitinib is limited by sunitinib-induced hypertension. Sunitinib 92-101 vascular endothelial growth factor A Rattus norvegicus 75-79 25275248-1 2014 OBJECTIVES: The therapeutic use of the vascular endothelial growth factor (VEGF) antagonist sunitinib is limited by sunitinib-induced hypertension. Sunitinib 116-125 vascular endothelial growth factor A Rattus norvegicus 39-73 25275248-1 2014 OBJECTIVES: The therapeutic use of the vascular endothelial growth factor (VEGF) antagonist sunitinib is limited by sunitinib-induced hypertension. Sunitinib 116-125 vascular endothelial growth factor A Rattus norvegicus 75-79 24802849-9 2014 The skin biopsies of mice administered sunitinib exhibited increased expression of Fas and FasL in the apoptotic keratinocytes in the epidermis. Sunitinib 39-48 Fas ligand (TNF superfamily, member 6) Mus musculus 91-95 25275248-8 2014 CONCLUSION: Our data indicate that early sunitinib-induced hypertension is associated with modest alterations in renal vascular function, but markedly increased renal sodium reabsorption, probably due to direct actions of the VEGF antagonist on the collecting duct, suggesting that VEGF receptors regulate renal Na+ absorption. Sunitinib 41-50 vascular endothelial growth factor A Rattus norvegicus 226-230 25275248-8 2014 CONCLUSION: Our data indicate that early sunitinib-induced hypertension is associated with modest alterations in renal vascular function, but markedly increased renal sodium reabsorption, probably due to direct actions of the VEGF antagonist on the collecting duct, suggesting that VEGF receptors regulate renal Na+ absorption. Sunitinib 41-50 vascular endothelial growth factor A Rattus norvegicus 282-286 24802849-10 2014 Our data revealed that Fas/FasL interaction mediates keratinocyte death in sunitinib-induced HFSR. Sunitinib 75-84 Fas ligand Homo sapiens 27-31 25089810-7 2014 Compound 54 is more potent than sunitinib not only against FLT3-WT AML cells but also active against sunitinib-resistant FLT3-ITD AML cells. Sunitinib 32-41 fms related receptor tyrosine kinase 3 Homo sapiens 59-63 25189542-5 2014 We observed in excess of 100 protein kinase targets plus various protein complexes involving, for instance, AMPK, TBK1 (sunitinib), and ILK (dasatinib). Sunitinib 120-129 TANK-binding kinase 1 Mus musculus 114-118 25547700-0 2014 [Genetic polymorphisms of PDGFR associated with thrombocytopenia in Chinese patients with clear-cell metastatic renal cell carcinoma treated with sunitinib]. Sunitinib 146-155 platelet derived growth factor receptor beta Homo sapiens 26-31 25547700-1 2014 OBJECTIVE: To explore the genetic polymorphisms of PDGFR associated with thrombocytopenia in Chinese patients with metastatic clear-cell renal cell carcinoma (mcc-RCC) treated with sunitinib. Sunitinib 181-190 platelet derived growth factor receptor beta Homo sapiens 51-56 25336977-9 2014 Among different VEGFR-TKIs, sorafenib and sunitinib had significant risk of death when compared with control arms, respectively. Sunitinib 42-51 kinase insert domain receptor Homo sapiens 16-21 25089810-7 2014 Compound 54 is more potent than sunitinib not only against FLT3-WT AML cells but also active against sunitinib-resistant FLT3-ITD AML cells. Sunitinib 32-41 fms related receptor tyrosine kinase 3 Homo sapiens 121-125 25089810-7 2014 Compound 54 is more potent than sunitinib not only against FLT3-WT AML cells but also active against sunitinib-resistant FLT3-ITD AML cells. Sunitinib 101-110 fms related receptor tyrosine kinase 3 Homo sapiens 121-125 25016061-5 2014 Assessment of tumoral neo-angiogenesis, assessed by morphometric analysis of the vascular network after CD31 immunolabeling, showed that both sunitinib and sorafenib reduced the microvessel area (-85% and -80%, respectively) and length (-80% and -78%, respectively) in treated compared to control tumors. Sunitinib 142-151 platelet and endothelial cell adhesion molecule 1 Rattus norvegicus 104-108 25100134-9 2014 CONCLUSIONS: Serum Ang-2 and MMP-2 and tumor HIF-1alpha were identified as relevant baseline biomarkers of sunitinib activity in advanced RCC, warranting further research into their prognostic versus predictive value. Sunitinib 107-116 angiopoietin 2 Homo sapiens 19-24 25100134-9 2014 CONCLUSIONS: Serum Ang-2 and MMP-2 and tumor HIF-1alpha were identified as relevant baseline biomarkers of sunitinib activity in advanced RCC, warranting further research into their prognostic versus predictive value. Sunitinib 107-116 matrix metallopeptidase 2 Homo sapiens 29-34 25016061-9 2014 While sunitinib induced a rapid (4h) and pronounced (5-fold) increase in caspase-3/7-dependent apoptosis, sorafenib seems to exert its cytotoxic activity through a different mechanism. Sunitinib 6-15 caspase 3 Rattus norvegicus 73-82 25228815-3 2014 In this study, we evaluate the changes in cell surface vascular endothelial growth factor receptor (VEGFR) expression on endothelial cells in culture treated with the antiangiogenic tyrosine kinase inhibitor drug sunitinib, using quantitative flow cytometry. Sunitinib 213-222 kinase insert domain receptor Homo sapiens 55-98 24612223-6 2014 Nilotinib, sunitinib, and imatinib were found to be potent CYP1A2 inhibitor. Sunitinib 11-20 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 59-65 25109654-6 2014 The results demonstrated that sunitinib was able to reverse the multidrug resistance of A549/DDP lung cancer cells, which was possibly associated with the downregulation of multidrug resistance-associated gene expression and the inhibition of AKT and ERK phosphorylation. Sunitinib 30-39 AKT serine/threonine kinase 1 Homo sapiens 243-246 25109654-6 2014 The results demonstrated that sunitinib was able to reverse the multidrug resistance of A549/DDP lung cancer cells, which was possibly associated with the downregulation of multidrug resistance-associated gene expression and the inhibition of AKT and ERK phosphorylation. Sunitinib 30-39 mitogen-activated protein kinase 1 Homo sapiens 251-254 24800947-4 2014 We review evidence to evaluate whether sunitinib may have value as re-challenge therapy in patients who have progressed on prior targeted therapy with sunitinib and/or an alternative tyrosine kinase inhibitor or mammalian target of rapamycin inhibitor. Sunitinib 39-48 mechanistic target of rapamycin kinase Homo sapiens 212-241 25228815-3 2014 In this study, we evaluate the changes in cell surface vascular endothelial growth factor receptor (VEGFR) expression on endothelial cells in culture treated with the antiangiogenic tyrosine kinase inhibitor drug sunitinib, using quantitative flow cytometry. Sunitinib 213-222 kinase insert domain receptor Homo sapiens 100-105 25228815-4 2014 We find that proangiogenic VEGFR2 cell surface receptor numbers are increased with sunitinib treatment. Sunitinib 83-92 kinase insert domain receptor Homo sapiens 27-33 24927771-1 2014 We performed a systematic review and meta-analysis of thyroid function abnormalities associated with seven vascular endothelial growth factor receptor (VEGFR) targeted tyrosine kinase inhibitors (sorafenib, sunitinib, axitinib, cediranib, pazopanib, regorafenib and vandetanib). Sunitinib 207-216 kinase insert domain receptor Homo sapiens 152-157 25200065-0 2014 Twist1 expression induced by sunitinib accelerates tumor cell vasculogenic mimicry by increasing the population of CD133+ cells in triple-negative breast cancer. Sunitinib 29-38 twist basic helix-loop-helix transcription factor 1 Mus musculus 0-6 25200065-0 2014 Twist1 expression induced by sunitinib accelerates tumor cell vasculogenic mimicry by increasing the population of CD133+ cells in triple-negative breast cancer. Sunitinib 29-38 prominin 1 Mus musculus 115-120 25193011-6 2014 He was subsequently treated with immunochemotherapy, tyrosine kinase inhibitors (TKIs), and mTOR inhibitors (mTORIs) - that is sunitinib, everolimus, and sorafenib. Sunitinib 127-136 mechanistic target of rapamycin kinase Homo sapiens 92-96 25145356-0 2014 Blocking autocrine VEGF signaling by sunitinib, an anti-cancer drug, promotes embryonic stem cell self-renewal and somatic cell reprogramming. Sunitinib 37-46 vascular endothelial growth factor A Mus musculus 19-23 25145356-3 2014 Blocking VEGF signaling with sunitinib, an anti-cancer drug and a receptor tyrosine kinase (RTK) inhibitor mainly targeting VEGF receptors (VEGFRs), is capable of maintaining the mESCs in the undifferentiated state without the need for feeder cells or LIF. Sunitinib 29-38 vascular endothelial growth factor A Mus musculus 9-13 25145356-3 2014 Blocking VEGF signaling with sunitinib, an anti-cancer drug and a receptor tyrosine kinase (RTK) inhibitor mainly targeting VEGF receptors (VEGFRs), is capable of maintaining the mESCs in the undifferentiated state without the need for feeder cells or LIF. Sunitinib 29-38 vascular endothelial growth factor A Mus musculus 124-128 25145356-3 2014 Blocking VEGF signaling with sunitinib, an anti-cancer drug and a receptor tyrosine kinase (RTK) inhibitor mainly targeting VEGF receptors (VEGFRs), is capable of maintaining the mESCs in the undifferentiated state without the need for feeder cells or LIF. Sunitinib 29-38 kinase insert domain protein receptor Mus musculus 140-146 25145356-3 2014 Blocking VEGF signaling with sunitinib, an anti-cancer drug and a receptor tyrosine kinase (RTK) inhibitor mainly targeting VEGF receptors (VEGFRs), is capable of maintaining the mESCs in the undifferentiated state without the need for feeder cells or LIF. Sunitinib 29-38 leukemia inhibitory factor Mus musculus 252-255 25145356-5 2014 Sunitinib also promotes iPSC generation from MEFs with only Oct4. Sunitinib 0-9 POU domain, class 5, transcription factor 1, related sequence 1 Mus musculus 60-64 25145356-6 2014 Knocking down VEGFR2 or blocking it with neutralizing antibody mimicks the effect of sunitinib, indicating that blocking VEGF/VEGFR signaling is indeed beneficial to the self-renewal of mESCs. Sunitinib 85-94 kinase insert domain protein receptor Mus musculus 14-20 25145356-6 2014 Knocking down VEGFR2 or blocking it with neutralizing antibody mimicks the effect of sunitinib, indicating that blocking VEGF/VEGFR signaling is indeed beneficial to the self-renewal of mESCs. Sunitinib 85-94 vascular endothelial growth factor A Mus musculus 14-18 25145356-11 2014 Blocking VEGF signaling with sunitinib or other small molecules help to maintain the mESCs in the ground state of pluripotency. Sunitinib 29-38 vascular endothelial growth factor A Mus musculus 9-13 25253994-3 2014 METHODS: PDGFR signaling was inhibited using Sunitinib malate, Imatinib mesylate or Regorafenib in murine and human luminal-like and claudin-low mammary tumor cell lines or Masitinib in only the human cell lines. Sunitinib 45-61 platelet derived growth factor receptor, beta polypeptide Mus musculus 9-14 25309777-4 2014 Vascular endothelial growth factor (VEGF)- and RET-directed therapies such as sorafenib, motesanib, and sunitinib have been shown to be the most effective at inducing clinical responses and stabilizing the disease process. Sunitinib 104-113 vascular endothelial growth factor A Homo sapiens 0-34 25309777-4 2014 Vascular endothelial growth factor (VEGF)- and RET-directed therapies such as sorafenib, motesanib, and sunitinib have been shown to be the most effective at inducing clinical responses and stabilizing the disease process. Sunitinib 104-113 vascular endothelial growth factor A Homo sapiens 36-40 25309777-4 2014 Vascular endothelial growth factor (VEGF)- and RET-directed therapies such as sorafenib, motesanib, and sunitinib have been shown to be the most effective at inducing clinical responses and stabilizing the disease process. Sunitinib 104-113 ret proto-oncogene Homo sapiens 47-50 24127298-11 2014 Treatment with VEGFR TKIs sorafenib, sunitinib and pazopanib is associated with a significant increase in the risk of GI events in patients with cancer, and frequent clinical monitoring should be emphasized when managing these three and newer VEGFR TKIs. Sunitinib 37-46 kinase insert domain receptor Homo sapiens 15-20 25068886-7 2014 Interestingly, sunitinib modulated tumor infiltration with bone marrow-derived cells (CD45+), recruitment of M2-like macrophages (CD163+) and activation of inflammatory pathways (phospho-STAT3) in a manner that was age-dependent. Sunitinib 15-24 signal transducer and activator of transcription 3 Mus musculus 187-192 24127298-11 2014 Treatment with VEGFR TKIs sorafenib, sunitinib and pazopanib is associated with a significant increase in the risk of GI events in patients with cancer, and frequent clinical monitoring should be emphasized when managing these three and newer VEGFR TKIs. Sunitinib 37-46 kinase insert domain receptor Homo sapiens 243-248 25017943-6 2014 Interestingly, pharmacological lipogenesis inhibition with orlistat or fatty acid synthase downregulation with shRNA inhibited tumor regrowth and metastases after sunitinib treatment withdrawal. Sunitinib 163-172 fatty acid synthase Homo sapiens 71-90 25101168-10 2014 RESULTS: The window chamber model was useful to demonstrate the inhibition of angiogenesis using the VEGF pathway targeted agent Sunitinib. Sunitinib 129-138 vascular endothelial growth factor A Mus musculus 101-105 24423438-4 2014 Vascular endothelial growth factor related upstream Ras, Erk1/2 and STAT1 phosphorylation activity in Caki-1 and NGAL transfected Caki-1 cells after sunitinib treatment was analyzed using Western blot. Sunitinib 149-158 lipocalin 2 Homo sapiens 113-117 24842548-1 2014 Our prior phase I study of the combination of vascular endothelial growth factor (VEGF) antibody, bevacizumab, and VEGF receptor (VEGFR) inhibitor, sunitinib, in advanced solid tumors identified an encouraging response evaluation. Sunitinib 148-157 kinase insert domain receptor Homo sapiens 115-128 24842548-1 2014 Our prior phase I study of the combination of vascular endothelial growth factor (VEGF) antibody, bevacizumab, and VEGF receptor (VEGFR) inhibitor, sunitinib, in advanced solid tumors identified an encouraging response evaluation. Sunitinib 148-157 kinase insert domain receptor Homo sapiens 130-135 24936559-4 2014 METHODS: We present a case report of a patient with a VIP-secreting malignant pheochromocytoma manifested as severe watery diarrhea, with an exquisite clinical response to sunitinib. Sunitinib 172-181 vasoactive intestinal peptide Homo sapiens 54-57 24936559-11 2014 CONCLUSION: To the best of our knowledge, this is the first report of a case of malignant VIP-producing pheochromocytoma that was responsive to sunitinib. Sunitinib 144-153 vasoactive intestinal peptide Homo sapiens 90-93 24423438-0 2014 NGAL can alternately mediate sunitinib resistance in renal cell carcinoma. Sunitinib 29-38 lipocalin 2 Homo sapiens 0-4 24423438-1 2014 PURPOSE: Serum NGAL is highly expressed in patients with advanced renal cancer treated with sunitinib. Sunitinib 92-101 lipocalin 2 Homo sapiens 15-19 24423438-2 2014 We investigated the role of NGAL in sunitinib resistance in renal cell carcinoma to identify potential tactics to overcome it. Sunitinib 36-45 lipocalin 2 Homo sapiens 28-32 24423438-3 2014 MATERIALS AND METHODS: NGAL expression was correlated with sunitinib sensitivity. Sunitinib 59-68 lipocalin 2 Homo sapiens 23-27 24423438-4 2014 Vascular endothelial growth factor related upstream Ras, Erk1/2 and STAT1 phosphorylation activity in Caki-1 and NGAL transfected Caki-1 cells after sunitinib treatment was analyzed using Western blot. Sunitinib 149-158 signal transducer and activator of transcription 1 Homo sapiens 68-73 24974736-7 2014 Sunitinib or LY294002, but not erlotinib, significantly inhibited the platelet-induced proliferation of osteosarcoma cells, indicating that PDGF released from platelets plays an important role in the proliferation of osteosarcomas by activating the PDGFR and then Akt. Sunitinib 0-9 platelet derived growth factor receptor beta Homo sapiens 249-254 24974736-7 2014 Sunitinib or LY294002, but not erlotinib, significantly inhibited the platelet-induced proliferation of osteosarcoma cells, indicating that PDGF released from platelets plays an important role in the proliferation of osteosarcomas by activating the PDGFR and then Akt. Sunitinib 0-9 AKT serine/threonine kinase 1 Homo sapiens 264-267 25085632-0 2014 Next generation sequencing analysis of platinum refractory advanced germ cell tumor sensitive to Sunitinib (Sutent ) a VEGFR2/PDGFRbeta/c-kit/ FLT3/RET/CSF1R inhibitor in a phase II trial. Sunitinib 97-106 kinase insert domain receptor Homo sapiens 119-125 25085632-0 2014 Next generation sequencing analysis of platinum refractory advanced germ cell tumor sensitive to Sunitinib (Sutent ) a VEGFR2/PDGFRbeta/c-kit/ FLT3/RET/CSF1R inhibitor in a phase II trial. Sunitinib 97-106 platelet derived growth factor receptor beta Homo sapiens 126-135 25085632-0 2014 Next generation sequencing analysis of platinum refractory advanced germ cell tumor sensitive to Sunitinib (Sutent ) a VEGFR2/PDGFRbeta/c-kit/ FLT3/RET/CSF1R inhibitor in a phase II trial. Sunitinib 97-106 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 136-141 25085632-0 2014 Next generation sequencing analysis of platinum refractory advanced germ cell tumor sensitive to Sunitinib (Sutent ) a VEGFR2/PDGFRbeta/c-kit/ FLT3/RET/CSF1R inhibitor in a phase II trial. Sunitinib 97-106 fms related receptor tyrosine kinase 3 Homo sapiens 143-147 25085632-0 2014 Next generation sequencing analysis of platinum refractory advanced germ cell tumor sensitive to Sunitinib (Sutent ) a VEGFR2/PDGFRbeta/c-kit/ FLT3/RET/CSF1R inhibitor in a phase II trial. Sunitinib 97-106 ret proto-oncogene Homo sapiens 148-151 25085632-0 2014 Next generation sequencing analysis of platinum refractory advanced germ cell tumor sensitive to Sunitinib (Sutent ) a VEGFR2/PDGFRbeta/c-kit/ FLT3/RET/CSF1R inhibitor in a phase II trial. Sunitinib 97-106 colony stimulating factor 1 receptor Homo sapiens 152-157 25085632-0 2014 Next generation sequencing analysis of platinum refractory advanced germ cell tumor sensitive to Sunitinib (Sutent ) a VEGFR2/PDGFRbeta/c-kit/ FLT3/RET/CSF1R inhibitor in a phase II trial. Sunitinib 108-114 platelet derived growth factor receptor beta Homo sapiens 126-135 25085632-4 2014 METHODS: Given the preclinical evidence implicating vascular endothelial growth factor (VEGF) signaling in the biology of germ cell tumors, we hypothesized that the vascular endothelial growth factor receptor (VEGFR) inhibitor sunitinib (Sutent) may possess important clinical activity in the treatment of this refractory disease. Sunitinib 227-236 vascular endothelial growth factor A Homo sapiens 52-86 25085632-4 2014 METHODS: Given the preclinical evidence implicating vascular endothelial growth factor (VEGF) signaling in the biology of germ cell tumors, we hypothesized that the vascular endothelial growth factor receptor (VEGFR) inhibitor sunitinib (Sutent) may possess important clinical activity in the treatment of this refractory disease. Sunitinib 227-236 kinase insert domain receptor Homo sapiens 165-208 25085632-4 2014 METHODS: Given the preclinical evidence implicating vascular endothelial growth factor (VEGF) signaling in the biology of germ cell tumors, we hypothesized that the vascular endothelial growth factor receptor (VEGFR) inhibitor sunitinib (Sutent) may possess important clinical activity in the treatment of this refractory disease. Sunitinib 227-236 kinase insert domain receptor Homo sapiens 210-215 25085632-4 2014 METHODS: Given the preclinical evidence implicating vascular endothelial growth factor (VEGF) signaling in the biology of germ cell tumors, we hypothesized that the vascular endothelial growth factor receptor (VEGFR) inhibitor sunitinib (Sutent) may possess important clinical activity in the treatment of this refractory disease. Sunitinib 238-244 kinase insert domain receptor Homo sapiens 165-208 25085632-4 2014 METHODS: Given the preclinical evidence implicating vascular endothelial growth factor (VEGF) signaling in the biology of germ cell tumors, we hypothesized that the vascular endothelial growth factor receptor (VEGFR) inhibitor sunitinib (Sutent) may possess important clinical activity in the treatment of this refractory disease. Sunitinib 238-244 kinase insert domain receptor Homo sapiens 210-215 25085632-9 2014 Next generation sequencing to understand this patient"s response to sunitinib revealed RET amplification, EGFR and KRAS amplification as relevant aberrations. Sunitinib 68-77 ret proto-oncogene Homo sapiens 87-90 25085632-9 2014 Next generation sequencing to understand this patient"s response to sunitinib revealed RET amplification, EGFR and KRAS amplification as relevant aberrations. Sunitinib 68-77 epidermal growth factor receptor Homo sapiens 106-110 25085632-9 2014 Next generation sequencing to understand this patient"s response to sunitinib revealed RET amplification, EGFR and KRAS amplification as relevant aberrations. Sunitinib 68-77 KRAS proto-oncogene, GTPase Homo sapiens 115-119 25085632-12 2014 Next generation sequencing of this "exceptional responder" identified the first reported case of a RET amplification as a potential basis of sensitivity to sunitinib (VEGFR2/PDGFRbeta/c-kit/ FLT3/RET/CSF1R inhibitor) in a patient with refractory germ cell tumor. Sunitinib 156-165 ret proto-oncogene Homo sapiens 99-102 25085632-12 2014 Next generation sequencing of this "exceptional responder" identified the first reported case of a RET amplification as a potential basis of sensitivity to sunitinib (VEGFR2/PDGFRbeta/c-kit/ FLT3/RET/CSF1R inhibitor) in a patient with refractory germ cell tumor. Sunitinib 156-165 kinase insert domain receptor Homo sapiens 167-173 25085632-12 2014 Next generation sequencing of this "exceptional responder" identified the first reported case of a RET amplification as a potential basis of sensitivity to sunitinib (VEGFR2/PDGFRbeta/c-kit/ FLT3/RET/CSF1R inhibitor) in a patient with refractory germ cell tumor. Sunitinib 156-165 platelet derived growth factor receptor beta Homo sapiens 174-183 25085632-12 2014 Next generation sequencing of this "exceptional responder" identified the first reported case of a RET amplification as a potential basis of sensitivity to sunitinib (VEGFR2/PDGFRbeta/c-kit/ FLT3/RET/CSF1R inhibitor) in a patient with refractory germ cell tumor. Sunitinib 156-165 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 184-189 25085632-12 2014 Next generation sequencing of this "exceptional responder" identified the first reported case of a RET amplification as a potential basis of sensitivity to sunitinib (VEGFR2/PDGFRbeta/c-kit/ FLT3/RET/CSF1R inhibitor) in a patient with refractory germ cell tumor. Sunitinib 156-165 fms related receptor tyrosine kinase 3 Homo sapiens 191-195 25085632-12 2014 Next generation sequencing of this "exceptional responder" identified the first reported case of a RET amplification as a potential basis of sensitivity to sunitinib (VEGFR2/PDGFRbeta/c-kit/ FLT3/RET/CSF1R inhibitor) in a patient with refractory germ cell tumor. Sunitinib 156-165 ret proto-oncogene Homo sapiens 196-199 25085632-12 2014 Next generation sequencing of this "exceptional responder" identified the first reported case of a RET amplification as a potential basis of sensitivity to sunitinib (VEGFR2/PDGFRbeta/c-kit/ FLT3/RET/CSF1R inhibitor) in a patient with refractory germ cell tumor. Sunitinib 156-165 colony stimulating factor 1 receptor Homo sapiens 200-205 25091988-7 2014 Low-dose sunitinib blocked the PDGF-BB-induced cell proliferation and PDGFR-beta signaling. Sunitinib 9-18 platelet derived growth factor receptor beta Homo sapiens 70-80 25091988-10 2014 CONCLUSIONS: Sunitinib mesylate inhibited the proliferation of primary human colonic fibroblasts through target-inhibited PDGFR signaling in vitro and in vivo. Sunitinib 13-22 platelet derived growth factor receptor beta Homo sapiens 122-127 24423438-5 2014 NGAL and vascular endothelial growth factor-A interaction with sunitinib therapeutic efficacy was monitored in renal cell carcinoma tumor xenografted mice by tumor growth inhibition, serum NGAL and vascular endothelial growth factor-a levels, and microscopic examination of tumor microvascular density. Sunitinib 63-72 vascular endothelial growth factor A Mus musculus 9-45 24423438-6 2014 RESULTS: Sunitinib cytotoxicity in various renal cell carcinoma cell lines was reversibly related to NGAL expression. Sunitinib 9-18 lipocalin 2 Homo sapiens 101-105 24423438-7 2014 Sunitinib showed the lowest 50% inhibitory concentration (5.53 muM) in Caki-1 cells, which had the lowest NGAL expression of these renal cell carcinoma cell lines. Sunitinib 0-9 lipocalin 2 Homo sapiens 106-110 24423438-8 2014 After sunitinib treatment adding NGAL inhibited Ras and Erk1/2 phosphorylation but activated STAT1alpha phosphorylation in Caki-1 cells and Caki-1 cells transfected with NGAL. Sunitinib 6-15 lipocalin 2 Homo sapiens 33-37 24423438-12 2014 CONCLUSIONS: NGAL in tumor cells may show crosstalk with vascular endothelial growth factor-a and alternative activation in stimulating tumor growth during sunitinib treatment. Sunitinib 156-165 lipocalin 2 Homo sapiens 13-17 25013907-0 2014 Association of toxicity of sorafenib and sunitinib for human keratinocytes with inhibition of signal transduction and activator of transcription 3 (STAT3). Sunitinib 41-50 signal transducer and activator of transcription 3 Homo sapiens 148-153 25130811-8 2014 The mRNA expression of C-MYC, hTERT and BCR-ABL was reduced significantly by SU11248 in a time-dependent manner (P < 0.05). Sunitinib 77-84 MYC proto-oncogene, bHLH transcription factor Homo sapiens 23-28 25130811-8 2014 The mRNA expression of C-MYC, hTERT and BCR-ABL was reduced significantly by SU11248 in a time-dependent manner (P < 0.05). Sunitinib 77-84 telomerase reverse transcriptase Homo sapiens 30-35 25130811-8 2014 The mRNA expression of C-MYC, hTERT and BCR-ABL was reduced significantly by SU11248 in a time-dependent manner (P < 0.05). Sunitinib 77-84 ABL proto-oncogene 1, non-receptor tyrosine kinase Homo sapiens 40-47 25130811-9 2014 Western blot detection showed that the expression of p-Akt protein in K562 cells decreased in dose-and time-dependent manner after SU11248 treatment, but the expression of Akt was not significantly changed. Sunitinib 131-138 AKT serine/threonine kinase 1 Homo sapiens 55-58 25130811-10 2014 It is concluded that SU11248 can inhibit the growth of K562 cells efficiently through inducing apoptosis, its mechanism may be closely relate with the expression down-regulation of C-MYC, hTERT, BCR-ABL and the inhibition of Akt phosphorylation. Sunitinib 21-28 MYC proto-oncogene, bHLH transcription factor Homo sapiens 181-186 25130811-10 2014 It is concluded that SU11248 can inhibit the growth of K562 cells efficiently through inducing apoptosis, its mechanism may be closely relate with the expression down-regulation of C-MYC, hTERT, BCR-ABL and the inhibition of Akt phosphorylation. Sunitinib 21-28 telomerase reverse transcriptase Homo sapiens 188-193 25130811-10 2014 It is concluded that SU11248 can inhibit the growth of K562 cells efficiently through inducing apoptosis, its mechanism may be closely relate with the expression down-regulation of C-MYC, hTERT, BCR-ABL and the inhibition of Akt phosphorylation. Sunitinib 21-28 ABL proto-oncogene 1, non-receptor tyrosine kinase Homo sapiens 195-202 25130811-10 2014 It is concluded that SU11248 can inhibit the growth of K562 cells efficiently through inducing apoptosis, its mechanism may be closely relate with the expression down-regulation of C-MYC, hTERT, BCR-ABL and the inhibition of Akt phosphorylation. Sunitinib 21-28 AKT serine/threonine kinase 1 Homo sapiens 225-228 25076341-4 2014 New therapies focus on specific molecular targets such as sunitinib, angiogenesis inhibitor, that target vascular endothelial growth factor receptor (VEGFR) and other growth factor receptors and everolimus, an inhibitor of the mammalian target of rapamycin. Sunitinib 58-67 kinase insert domain receptor Homo sapiens 105-148 25076341-4 2014 New therapies focus on specific molecular targets such as sunitinib, angiogenesis inhibitor, that target vascular endothelial growth factor receptor (VEGFR) and other growth factor receptors and everolimus, an inhibitor of the mammalian target of rapamycin. Sunitinib 58-67 kinase insert domain receptor Homo sapiens 150-155 25013907-2 2014 This study aimed to examine whether the toxicity of sorafenib and sunitinib for human keratinocytes was associated with inhibiting signal transduction and activator of transcription 3 (STAT3). Sunitinib 66-75 signal transducer and activator of transcription 3 Homo sapiens 185-190 25013907-3 2014 We studied whether STAT3 activity affects sorafenib- and sunitinib-induced cell growth inhibition in HaCaT cells by WST-8 assay. Sunitinib 57-66 signal transducer and activator of transcription 3 Homo sapiens 19-24 25013907-5 2014 HaCaT cells transfected with constitutively-active STAT3 (STAT3C) were resistant to the sorafenib- and sunitinib-induced cell growth inhibition. Sunitinib 103-112 signal transducer and activator of transcription 3 Homo sapiens 51-56 25013907-5 2014 HaCaT cells transfected with constitutively-active STAT3 (STAT3C) were resistant to the sorafenib- and sunitinib-induced cell growth inhibition. Sunitinib 103-112 signal transducer and activator of transcription 3 Homo sapiens 58-64 25013907-6 2014 STAT3 activity decreased after short-term treatment with sorafenib and sunitinib in a dose-dependent manner and recovered after long-term treatment with sorafenib and sunitinib at low doses. Sunitinib 71-80 signal transducer and activator of transcription 3 Homo sapiens 0-5 25013907-6 2014 STAT3 activity decreased after short-term treatment with sorafenib and sunitinib in a dose-dependent manner and recovered after long-term treatment with sorafenib and sunitinib at low doses. Sunitinib 167-176 signal transducer and activator of transcription 3 Homo sapiens 0-5 25013907-7 2014 Moreover, the expression of survivin and bcl-2 decreased after treatment with sorafenib and sunitinib was concomitant with variations in STAT3 activity. Sunitinib 92-101 BCL2 apoptosis regulator Homo sapiens 41-46 25013907-8 2014 Sorafenib-induced STAT3 inhibition was mediated by regulation via MAPK pathways in HaCaT cells, while sunitinib-induced STAT3 inhibition was not. Sunitinib 102-111 signal transducer and activator of transcription 3 Homo sapiens 120-125 25013907-9 2014 Thus, STAT3 activation mediating apoptosis suppressors may be a key factor in sorafenib and sunitinib-induced keratinocyte cytotoxicity. Sunitinib 92-101 signal transducer and activator of transcription 3 Homo sapiens 6-11 24482243-3 2014 Encouragingly, VEGF pathway targeted drugs such as bevacizumab, sunitinib and aflibercept have shown activity in certain settings. Sunitinib 64-73 vascular endothelial growth factor A Homo sapiens 15-19 24566734-5 2014 From univariate analysis, we found that the SNPs in CYP3A4, CYP3A5, and ABCB1 were associated with the clearance of both sunitinib and SU12662. Sunitinib 121-130 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 52-58 24215209-0 2014 Prognostic impact of baseline serum C-reactive protein in patients with metastatic renal cell carcinoma (RCC) treated with sunitinib. Sunitinib 123-132 C-reactive protein Homo sapiens 36-54 24215209-1 2014 OBJECTIVE: To evaluate the impact of baseline serum C-reactive protein (CRP) level on outcome in patients with metastatic renal cell carcinoma (mRCC) treated with sunitinib. Sunitinib 163-172 C-reactive protein Homo sapiens 52-70 24215209-1 2014 OBJECTIVE: To evaluate the impact of baseline serum C-reactive protein (CRP) level on outcome in patients with metastatic renal cell carcinoma (mRCC) treated with sunitinib. Sunitinib 163-172 C-reactive protein Homo sapiens 72-75 24215209-12 2014 CONCLUSION: Baseline serum CRP level is a strong independent variable linked with RR, PFS and OS in patients with mRCC treated with sunitinib. Sunitinib 132-141 C-reactive protein Homo sapiens 27-30 24566734-5 2014 From univariate analysis, we found that the SNPs in CYP3A4, CYP3A5, and ABCB1 were associated with the clearance of both sunitinib and SU12662. Sunitinib 121-130 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 60-66 24566734-5 2014 From univariate analysis, we found that the SNPs in CYP3A4, CYP3A5, and ABCB1 were associated with the clearance of both sunitinib and SU12662. Sunitinib 121-130 ATP binding cassette subfamily B member 1 Homo sapiens 72-77 24751611-7 2014 Sunitinib induced autophagy via upregulating beclin-1 expression which was blocked by chloroquine as evidenced by accumulated SQTSM1/p62 level. Sunitinib 0-9 beclin 1 Homo sapiens 45-53 24562646-7 2014 Immunofluorescence staining and an enzyme-linked immunosorbent assay confirmed that sunitinib-induced changes in tumor uptake of CF680R-3PRGD2 and CF750-BevF(ab")2 were correlated with changes in the levels of integrin alphavbeta3 and VEGF. Sunitinib 84-93 vascular endothelial growth factor A Mus musculus 235-239 24976727-11 2014 Cells treated with sorafenib or sunitinib expressed less interleukin-6 mRNA as well as less collagen type 1 mRNA and protein. Sunitinib 32-41 interleukin 6 Rattus norvegicus 57-70 24510251-4 2014 Two patients were treated with sunitinib at a standard dose (50 mg daily; 4 weeks on, 2 weeks off) after cyclophosphamide/vinblastine/dacarbazine chemotherapy, because vascular endothelial growth factor (VEGF)-positive cells were partly observed by immunohistochemical staining. Sunitinib 31-40 vascular endothelial growth factor A Homo sapiens 168-202 24510251-10 2014 In conclusion, sunitinib was effective in the treatment of malignant pheochromocytoma when VEGF-positive cells were observed in the tumor specimens. Sunitinib 15-24 vascular endothelial growth factor A Homo sapiens 91-95 24680659-3 2014 RESULTS: The baseline level of MMP-9 in nonresponders to sunitinib was significantly higher than that in responders, whereas the baseline level of TIMP-2 in nonresponders was significantly lower than that in responders. Sunitinib 57-66 matrix metallopeptidase 9 Homo sapiens 31-36 24680659-8 2014 CONCLUSIONS: An imbalance between the serum MMP-9 and TIMP-2 levels could be a novel biomarker to predict disease progression in patients with mRCC under treatment with sunitinib. Sunitinib 169-178 matrix metallopeptidase 9 Homo sapiens 44-49 24680659-8 2014 CONCLUSIONS: An imbalance between the serum MMP-9 and TIMP-2 levels could be a novel biomarker to predict disease progression in patients with mRCC under treatment with sunitinib. Sunitinib 169-178 TIMP metallopeptidase inhibitor 2 Homo sapiens 54-60 24751611-7 2014 Sunitinib induced autophagy via upregulating beclin-1 expression which was blocked by chloroquine as evidenced by accumulated SQTSM1/p62 level. Sunitinib 0-9 nucleoporin 62 Homo sapiens 133-136 24751611-8 2014 Furthermore, chloroquine augmented sunitinib-induced apoptosis by decreasing survivin level and increasing caspase 3 activity. Sunitinib 35-44 caspase 3 Homo sapiens 107-116 24751611-9 2014 Chloroquine also enhanced the antiangiogenic capacity of sunitinib as indicated by decreased CD34 expression and peritoneal/skin angiogenesis. Sunitinib 57-66 CD34 molecule Homo sapiens 93-97 24185817-0 2014 Sunitinib--CLIO conjugate: a VEGFR/PDGFR-targeting active MR probe. Sunitinib 0-9 kinase insert domain receptor Homo sapiens 29-34 24703573-13 2014 Among putative sunitinib targets, only RET was expressed and activated in analysed samples. Sunitinib 15-24 ret proto-oncogene Homo sapiens 39-42 25058995-1 2014 BACKGROUND: The VEGFR/PDGFR inhibitor sunitinib was approved in Israel in 2008 for the treatment of metastatic renal cell carcinoma (mRCC), based on an international trial. Sunitinib 38-47 kinase insert domain receptor Homo sapiens 16-21 25058995-1 2014 BACKGROUND: The VEGFR/PDGFR inhibitor sunitinib was approved in Israel in 2008 for the treatment of metastatic renal cell carcinoma (mRCC), based on an international trial. Sunitinib 38-47 platelet derived growth factor receptor beta Homo sapiens 22-27 24417304-1 2014 WHAT IS KNOWN AND OBJECTIVE: Sunitinib, a CYP3A4 substrate, is standard of care treatment in metastatic renal cell carcinoma (mRCC) and is administered orally as a single dose of 50 mg, in a 4 weeks on/2 weeks off regimen. Sunitinib 29-38 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 42-48 24417304-16 2014 Due to adverse drug reactions and comorbidity, patients under sunitinib, a CYP3A4 substrate, took an average of 6 8 comedications provoking an important risk of major-to-moderate drug-drug interactions. Sunitinib 62-71 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 75-81 24185817-0 2014 Sunitinib--CLIO conjugate: a VEGFR/PDGFR-targeting active MR probe. Sunitinib 0-9 platelet derived growth factor receptor beta Homo sapiens 35-40 24185817-1 2014 PURPOSE: This study was conducted to evaluate feasibility of sunitinib-CLIO conjugate as a vascular endothelial growth factor receptor/platelet-derived growth factor receptor (VEGFR/PDGFR)-specific magnetic resonance (MR) probe. Sunitinib 61-70 kinase insert domain receptor Homo sapiens 176-181 24185817-1 2014 PURPOSE: This study was conducted to evaluate feasibility of sunitinib-CLIO conjugate as a vascular endothelial growth factor receptor/platelet-derived growth factor receptor (VEGFR/PDGFR)-specific magnetic resonance (MR) probe. Sunitinib 61-70 platelet derived growth factor receptor beta Homo sapiens 182-187 24185817-2 2014 PROCEDURE: VEGFR/PDGFR-targeting MR probe was synthesized by conjugating cross-linked iron-oxide (CLIO) with tyrosine-kinase inhibitor (sunitinib). Sunitinib 136-145 kinase insert domain receptor Homo sapiens 11-16 24185817-2 2014 PROCEDURE: VEGFR/PDGFR-targeting MR probe was synthesized by conjugating cross-linked iron-oxide (CLIO) with tyrosine-kinase inhibitor (sunitinib). Sunitinib 136-145 platelet derived growth factor receptor beta Homo sapiens 17-22 24185817-9 2014 CONCLUSIONS: Sunitinib-CLIO conjugate can be used as an active MR probe for quantifying VEGFR/PDGFR. Sunitinib 13-22 kinase insert domain receptor Homo sapiens 88-93 24185817-9 2014 CONCLUSIONS: Sunitinib-CLIO conjugate can be used as an active MR probe for quantifying VEGFR/PDGFR. Sunitinib 13-22 platelet derived growth factor receptor beta Homo sapiens 94-99 24825438-0 2014 The effect of ABCG2 genotype on the population pharmacokinetics of sunitinib in patients with renal cell carcinoma. Sunitinib 67-76 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 14-19 24723451-1 2014 Sunitinib, an inhibitor of kinases, including VEGFR and platelet-derived growth factor receptor (PDGFR), efficiently induces apoptosis in vitro in glioblastoma (GBM) cells, but does not show any survival benefit in vivo. Sunitinib 0-9 kinase insert domain receptor Homo sapiens 46-51 24723451-1 2014 Sunitinib, an inhibitor of kinases, including VEGFR and platelet-derived growth factor receptor (PDGFR), efficiently induces apoptosis in vitro in glioblastoma (GBM) cells, but does not show any survival benefit in vivo. Sunitinib 0-9 platelet derived growth factor receptor beta Homo sapiens 56-95 24723451-1 2014 Sunitinib, an inhibitor of kinases, including VEGFR and platelet-derived growth factor receptor (PDGFR), efficiently induces apoptosis in vitro in glioblastoma (GBM) cells, but does not show any survival benefit in vivo. Sunitinib 0-9 platelet derived growth factor receptor beta Homo sapiens 97-102 24825438-15 2014 CONCLUSIONS: This analysis provides a population PK model of sunitinib with the ABCG2 421C>A genotype as a predictive covariate for CL/F. Sunitinib 61-70 crooked neck pre-mRNA splicing factor 1 Homo sapiens 135-139 24825438-16 2014 It also suggests that IOV and change of CL/F over time need to be considered to predict the sunitinib PK more accurately. Sunitinib 92-101 crooked neck pre-mRNA splicing factor 1 Homo sapiens 40-44 24825438-15 2014 CONCLUSIONS: This analysis provides a population PK model of sunitinib with the ABCG2 421C>A genotype as a predictive covariate for CL/F. Sunitinib 61-70 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 80-85 24877032-0 2014 Post-treatment neutrophil-to-lymphocyte ratio in predicting prognosis in patients with metastatic clear cell renal cell carcinoma receiving sunitinib as first line therapy. Sunitinib 140-149 solute carrier family 35 member G1 Homo sapiens 0-4 24883179-4 2014 Early FLT3 inhibitors (including sunitinib, midostaurin, and lestaurtinib) demonstrated significant promise in preclinical models of FLT3 mutant AML. Sunitinib 33-42 fms related receptor tyrosine kinase 3 Homo sapiens 6-10 24883179-4 2014 Early FLT3 inhibitors (including sunitinib, midostaurin, and lestaurtinib) demonstrated significant promise in preclinical models of FLT3 mutant AML. Sunitinib 33-42 fms related receptor tyrosine kinase 3 Homo sapiens 133-137 24914410-6 2014 The effect of sunitinib on Notch-1 expression was determined by Western blot in cultured MDA-MB-468 cells. Sunitinib 14-23 notch receptor 1 Homo sapiens 27-34 24914410-13 2014 Sunitinib significantly increased the expression of Notch-1 protein in cultured MDA-MB-468 or MDA-MB-231 cells. Sunitinib 0-9 notch receptor 1 Homo sapiens 52-59 24914410-15 2014 These findings support the hypothesis that the possibility should be considered of sunitinib increasing breast CSCs though it inhibits TNBC tumor angiogenesis and growth/progression, and that effects of sunitinib on Notch expression and hypoxia may increase breast cancer stem cells. Sunitinib 203-212 notch receptor 1 Homo sapiens 216-221 24819355-1 2014 We have investigated in vitro the metabolic capability of 3 extrahepatic cytochromes P-450, CYP1A1, 1B1 and 2J2, known to be over-expressed in various tumors, to biotransform 5 tyrosine kinase inhibitors (TKI): dasatinib, imatinib, nilotinib, sorafenib and sunitinib. Sunitinib 257-266 cytochrome P450 family 1 subfamily A member 1 Homo sapiens 92-103 24786599-9 2014 CONCLUSIONS: Our results suggest that the expression of phosphorylated (i.e., activated) KDR in tumour stroma might be used as predictive biomarker for the clinical outcome in renal cell carcinoma first-line sunitinib-treated patients. Sunitinib 208-217 kinase insert domain receptor Homo sapiens 89-92 24045439-8 2014 Similarly, sunitinib, a small-molecule inhibitor of RET, blocked GDNF-mediated activation of ERK and AKT. Sunitinib 11-20 ret proto-oncogene Homo sapiens 52-55 24310825-3 2014 METHODS: We report four pediatric patients with various malignancies (chronic myelogenous leukemia, acute lymphoblastic leukemia, and glioma/renal cell carcinoma) who developed nephrotic syndrome during treatment with TK inhibitors (imatinib, sunitinib, dasatinib, and quizartinib). Sunitinib 243-252 TXK tyrosine kinase Homo sapiens 218-220 23728343-3 2014 We found that the multi-kinase inhibitors midostaurin and sunitinib downregulate MITF protein levels. Sunitinib 58-67 melanocyte inducing transcription factor Homo sapiens 81-85 24816095-6 2014 Sunitinib forms nine preserved bond paths corresponding to hydrogen bonds and also to the C-H O and C-H pi contacts common to the VEGRF2, CDK2, G2, KIT and IT kinases. Sunitinib 0-9 cyclin dependent kinase 2 Homo sapiens 142-146 24289201-1 2014 OBJECTIVES: The purpose of this study was to confirm the inhibitory effects of sunitinib, an angiogenesis inhibitor that targets tyrosine kinases of vascular endothelial growth factor receptor (VEGFR) family and platelet-derived growth factor receptor (PDGFR) family, on arthritis in mice with type II collagen-induced arthritis (CIA). Sunitinib 79-88 platelet derived growth factor receptor, beta polypeptide Mus musculus 212-251 24289201-1 2014 OBJECTIVES: The purpose of this study was to confirm the inhibitory effects of sunitinib, an angiogenesis inhibitor that targets tyrosine kinases of vascular endothelial growth factor receptor (VEGFR) family and platelet-derived growth factor receptor (PDGFR) family, on arthritis in mice with type II collagen-induced arthritis (CIA). Sunitinib 79-88 platelet derived growth factor receptor, beta polypeptide Mus musculus 253-258 24289201-1 2014 OBJECTIVES: The purpose of this study was to confirm the inhibitory effects of sunitinib, an angiogenesis inhibitor that targets tyrosine kinases of vascular endothelial growth factor receptor (VEGFR) family and platelet-derived growth factor receptor (PDGFR) family, on arthritis in mice with type II collagen-induced arthritis (CIA). Sunitinib 79-88 nuclear receptor coactivator 5 Mus musculus 294-334 24759734-0 2014 Sunitinib suppress neuroblastoma growth through degradation of MYCN and inhibition of angiogenesis. Sunitinib 0-9 MYCN proto-oncogene, bHLH transcription factor Homo sapiens 63-67 24759734-7 2014 Treatment with sunitinib decreased MYCN protein levels by inhibition of PI3K/AKT signaling and GSK3beta. Sunitinib 15-24 MYCN proto-oncogene, bHLH transcription factor Homo sapiens 35-39 24759734-7 2014 Treatment with sunitinib decreased MYCN protein levels by inhibition of PI3K/AKT signaling and GSK3beta. Sunitinib 15-24 glycogen synthase kinase 3 beta Homo sapiens 95-103 24759734-9 2014 Sunitinib significantly inhibited the growth of established, subcutaneous MYCN-amplified neuroblastoma xenografts in nude mice and demonstrated an anti-angiogenic effect in vivo with a reduction of tumor vasculature and a decrease of MYCN expression. Sunitinib 0-9 v-myc avian myelocytomatosis viral related oncogene, neuroblastoma derived Mus musculus 74-78 24759734-9 2014 Sunitinib significantly inhibited the growth of established, subcutaneous MYCN-amplified neuroblastoma xenografts in nude mice and demonstrated an anti-angiogenic effect in vivo with a reduction of tumor vasculature and a decrease of MYCN expression. Sunitinib 0-9 v-myc avian myelocytomatosis viral related oncogene, neuroblastoma derived Mus musculus 234-238 24045439-8 2014 Similarly, sunitinib, a small-molecule inhibitor of RET, blocked GDNF-mediated activation of ERK and AKT. Sunitinib 11-20 glial cell derived neurotrophic factor Homo sapiens 65-69 24783183-2 2014 Several recent findings that there are activating mutations in the KIT and PDGFRA (platelet-derived growth factor receptor-alpha) genes of GISTs provide the rationale for using targeted therapies such as imatinib or sunitinib. Sunitinib 216-225 platelet derived growth factor receptor alpha Homo sapiens 83-128 24783183-3 2014 Sunitinib, an oral multitargeted receptor tyrosine kinase inhibitor that inhibits kinases such as KIT, PDGFR (platelet-derived growth factor recepter), and VEGFR (vascular endothelial growth factor receptor), was recently approved for the treatment of imatinib-refractory GIST. Sunitinib 0-9 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 98-101 24783183-2 2014 Several recent findings that there are activating mutations in the KIT and PDGFRA (platelet-derived growth factor receptor-alpha) genes of GISTs provide the rationale for using targeted therapies such as imatinib or sunitinib. Sunitinib 216-225 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 67-70 24783183-2 2014 Several recent findings that there are activating mutations in the KIT and PDGFRA (platelet-derived growth factor receptor-alpha) genes of GISTs provide the rationale for using targeted therapies such as imatinib or sunitinib. Sunitinib 216-225 platelet derived growth factor receptor alpha Homo sapiens 75-81 24045439-8 2014 Similarly, sunitinib, a small-molecule inhibitor of RET, blocked GDNF-mediated activation of ERK and AKT. Sunitinib 11-20 mitogen-activated protein kinase 1 Homo sapiens 93-96 24783183-3 2014 Sunitinib, an oral multitargeted receptor tyrosine kinase inhibitor that inhibits kinases such as KIT, PDGFR (platelet-derived growth factor recepter), and VEGFR (vascular endothelial growth factor receptor), was recently approved for the treatment of imatinib-refractory GIST. Sunitinib 0-9 platelet derived growth factor receptor beta Homo sapiens 103-108 24783183-3 2014 Sunitinib, an oral multitargeted receptor tyrosine kinase inhibitor that inhibits kinases such as KIT, PDGFR (platelet-derived growth factor recepter), and VEGFR (vascular endothelial growth factor receptor), was recently approved for the treatment of imatinib-refractory GIST. Sunitinib 0-9 platelet derived growth factor receptor beta Homo sapiens 110-149 24045439-8 2014 Similarly, sunitinib, a small-molecule inhibitor of RET, blocked GDNF-mediated activation of ERK and AKT. Sunitinib 11-20 AKT serine/threonine kinase 1 Homo sapiens 101-104 24783183-3 2014 Sunitinib, an oral multitargeted receptor tyrosine kinase inhibitor that inhibits kinases such as KIT, PDGFR (platelet-derived growth factor recepter), and VEGFR (vascular endothelial growth factor receptor), was recently approved for the treatment of imatinib-refractory GIST. Sunitinib 0-9 kinase insert domain receptor Homo sapiens 156-161 24045439-9 2014 Inhibition of RET either by gene knockdown or by treatment with sunitinib or vandetanib reduced RET-dependent growth of luminal breast cancer cells. Sunitinib 64-73 ret proto-oncogene Homo sapiens 14-17 24783183-3 2014 Sunitinib, an oral multitargeted receptor tyrosine kinase inhibitor that inhibits kinases such as KIT, PDGFR (platelet-derived growth factor recepter), and VEGFR (vascular endothelial growth factor receptor), was recently approved for the treatment of imatinib-refractory GIST. Sunitinib 0-9 kinase insert domain receptor Homo sapiens 163-206 24045439-9 2014 Inhibition of RET either by gene knockdown or by treatment with sunitinib or vandetanib reduced RET-dependent growth of luminal breast cancer cells. Sunitinib 64-73 ret proto-oncogene Homo sapiens 96-99 24045439-11 2014 Parallel experiments using treatment with tamoxifen and sunitinib confirmed the increased effectiveness of dual inhibition of the ER and RET pathways in regulating cell growth. Sunitinib 56-65 ret proto-oncogene Homo sapiens 137-140 24118906-0 2014 The evaluation of efficacy and safety of sunitinib on EGFR-TKI pretreated advanced non-small cell lung cancer patients in China. Sunitinib 41-50 epidermal growth factor receptor Homo sapiens 54-58 24612599-3 2014 This study aimed to assess the association between initial tumor size, involved organs, pre-treatment C-reactive protein (CRP) levels, and reduction in tumor size in patients with clear cell RCC (CCRCC) treated with sunitinib. Sunitinib 216-225 C-reactive protein Homo sapiens 102-120 24425345-3 2014 Sunitinib not only inhibits PDGFRs, KIT, and FLT3, it also blocks VEGFRs and thus serves as an antiangiogenic agent. Sunitinib 0-9 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 36-39 24425345-3 2014 Sunitinib not only inhibits PDGFRs, KIT, and FLT3, it also blocks VEGFRs and thus serves as an antiangiogenic agent. Sunitinib 0-9 fms related receptor tyrosine kinase 3 Homo sapiens 45-49 24704536-6 2014 Plasma Ang2 decreased with sunitinib treatment and increased at time of disease progression. Sunitinib 27-36 angiopoietin 2 Homo sapiens 7-11 24704536-7 2014 In the RCC mouse, dual Ang1/2 and Ang2 inhibition improved the activity of sunitinib. Sunitinib 75-84 angiopoietin 1 Mus musculus 23-29 24704536-7 2014 In the RCC mouse, dual Ang1/2 and Ang2 inhibition improved the activity of sunitinib. Sunitinib 75-84 angiopoietin 2 Homo sapiens 34-38 24704536-8 2014 Combined Ang1/2 and VEGFR inhibition prevented the resumption of blood flow associated with sunitinib resistance. Sunitinib 92-101 angiopoietin 1 Homo sapiens 9-15 24704536-8 2014 Combined Ang1/2 and VEGFR inhibition prevented the resumption of blood flow associated with sunitinib resistance. Sunitinib 92-101 kinase insert domain receptor Homo sapiens 20-25 24704536-9 2014 Thus, Ang2 inhibition, independent of Ang1 inhibition, improves the activity of sunitinib and plasma Ang2 increases in the setting of progression on sunitinib possibly contributing to resistance. Sunitinib 80-89 angiopoietin 2 Homo sapiens 6-10 24704536-9 2014 Thus, Ang2 inhibition, independent of Ang1 inhibition, improves the activity of sunitinib and plasma Ang2 increases in the setting of progression on sunitinib possibly contributing to resistance. Sunitinib 149-158 angiopoietin 2 Homo sapiens 6-10 24704536-9 2014 Thus, Ang2 inhibition, independent of Ang1 inhibition, improves the activity of sunitinib and plasma Ang2 increases in the setting of progression on sunitinib possibly contributing to resistance. Sunitinib 149-158 angiopoietin 2 Homo sapiens 101-105 24742865-9 2014 Sunitinib plus rapamycin markedly induced versican, IDO, arginase 1, IL-6, and TGF-beta expression in the lungs, whereas it reduced IDO and IL-10 expression in the primary tumor tissues. Sunitinib 0-9 versican Mus musculus 42-50 24742865-9 2014 Sunitinib plus rapamycin markedly induced versican, IDO, arginase 1, IL-6, and TGF-beta expression in the lungs, whereas it reduced IDO and IL-10 expression in the primary tumor tissues. Sunitinib 0-9 indoleamine 2,3-dioxygenase 1 Mus musculus 52-55 24742865-9 2014 Sunitinib plus rapamycin markedly induced versican, IDO, arginase 1, IL-6, and TGF-beta expression in the lungs, whereas it reduced IDO and IL-10 expression in the primary tumor tissues. Sunitinib 0-9 arginase, liver Mus musculus 57-67 24742865-9 2014 Sunitinib plus rapamycin markedly induced versican, IDO, arginase 1, IL-6, and TGF-beta expression in the lungs, whereas it reduced IDO and IL-10 expression in the primary tumor tissues. Sunitinib 0-9 interleukin 6 Mus musculus 69-73 24742865-9 2014 Sunitinib plus rapamycin markedly induced versican, IDO, arginase 1, IL-6, and TGF-beta expression in the lungs, whereas it reduced IDO and IL-10 expression in the primary tumor tissues. Sunitinib 0-9 transforming growth factor, beta 1 Mus musculus 79-87 24742865-9 2014 Sunitinib plus rapamycin markedly induced versican, IDO, arginase 1, IL-6, and TGF-beta expression in the lungs, whereas it reduced IDO and IL-10 expression in the primary tumor tissues. Sunitinib 0-9 indoleamine 2,3-dioxygenase 1 Mus musculus 132-135 24742865-9 2014 Sunitinib plus rapamycin markedly induced versican, IDO, arginase 1, IL-6, and TGF-beta expression in the lungs, whereas it reduced IDO and IL-10 expression in the primary tumor tissues. Sunitinib 0-9 interleukin 10 Mus musculus 140-145 24744988-12 2014 Similarly, several marketed tyrosine kinase inhibitors (TKIs) in the US (sorafenib, sunitinib, vandetanib, cabozantinib, regorafenib) are potent RET inhibitors in vitro. Sunitinib 84-93 ret proto-oncogene Homo sapiens 145-148 24105638-4 2014 Mice immunized with recombinant alpha-lactalbumin, a lactation protein expressed on majority of breast tumors were treated with 1 mg of Sunitinib for seven consecutive days beginning (1) concurrently, on the day of alpha-lactalbumin immunization or (2) sequentially, on day 9 after immunization. Sunitinib 136-145 lactalbumin, alpha Mus musculus 32-49 24105638-5 2014 Ten-day lymph nodes or 21 day spleens were tested by ELISPOT assays and flow cytometry to evaluate responsiveness to alpha-lactalbumin immunization in presence of Sunitinib and distribution of cells involved in T cell antigen priming and proliferation in different lymphoid compartments. Sunitinib 163-172 lactalbumin alpha Homo sapiens 117-134 24105638-7 2014 Our studies reveal that concurrent administration of Sunitinib with active vaccination against a targeted tumor antigen inhibits priming to the immunogen due to a drastic decrease in CD11b+CD11c+ antigen presenting cells, leading to failure of vaccination. Sunitinib 53-62 integrin subunit alpha M Homo sapiens 183-188 24105638-7 2014 Our studies reveal that concurrent administration of Sunitinib with active vaccination against a targeted tumor antigen inhibits priming to the immunogen due to a drastic decrease in CD11b+CD11c+ antigen presenting cells, leading to failure of vaccination. Sunitinib 53-62 integrin subunit alpha X Homo sapiens 189-194 24612599-3 2014 This study aimed to assess the association between initial tumor size, involved organs, pre-treatment C-reactive protein (CRP) levels, and reduction in tumor size in patients with clear cell RCC (CCRCC) treated with sunitinib. Sunitinib 216-225 C-reactive protein Homo sapiens 122-125 24612599-11 2014 CONCLUSIONS: Patients with CCRCC with smaller lung metastatic lesions and lower CRP levels may achieve greater percent reductions in tumor size with sunitinib therapy than patients with extra-pulmonary lesions, large lung lesions, and/or higher CRP levels. Sunitinib 149-158 C-reactive protein Homo sapiens 80-83 23975598-5 2014 We found that PDGFR-beta expression in cell seems to be a protective factor against Sunitinib resistance formation. Sunitinib 84-93 platelet derived growth factor receptor beta Homo sapiens 14-24 24606768-1 2014 BACKGROUND: This phase II study evaluated the efficacy and safety/tolerability of sunitinib plus trastuzumab in patients with HER2-positive advanced breast cancer (ABC). Sunitinib 82-91 erb-b2 receptor tyrosine kinase 2 Homo sapiens 126-130 24606768-15 2014 CONCLUSIONS: Sunitinib plus trastuzumab demonstrated antitumor activity in patients with HER2-positive ABC, particularly those who were treatment-naive or had only received prior adjuvant treatment. Sunitinib 13-22 erb-b2 receptor tyrosine kinase 2 Homo sapiens 89-93 24606768-16 2014 Sunitinib plus trastuzumab had acceptable safety and tolerability in patients with HER2-positive ABC who had not received prior anthracycline therapy. Sunitinib 0-9 erb-b2 receptor tyrosine kinase 2 Homo sapiens 83-87 24247421-0 2014 Development of cardiac hypertrophy by sunitinib in vivo and in vitro rat cardiomyocytes is influenced by the aryl hydrocarbon receptor signaling pathway. Sunitinib 38-47 aryl hydrocarbon receptor Rattus norvegicus 109-134 23975598-6 2014 In addition, we found that both SK1 and ERK were activated in Sunitinib-resistance cell lines and SK1 and ERK inhibitors could resensitize Sunitinib-resistant cell lines. Sunitinib 62-71 sphingosine kinase 1 Homo sapiens 32-35 23975598-6 2014 In addition, we found that both SK1 and ERK were activated in Sunitinib-resistance cell lines and SK1 and ERK inhibitors could resensitize Sunitinib-resistant cell lines. Sunitinib 62-71 EPH receptor B2 Homo sapiens 40-43 23975598-6 2014 In addition, we found that both SK1 and ERK were activated in Sunitinib-resistance cell lines and SK1 and ERK inhibitors could resensitize Sunitinib-resistant cell lines. Sunitinib 139-148 sphingosine kinase 1 Homo sapiens 32-35 24414551-1 2014 BACKGROUND: Sunitinib treatment results in a compensatory increase in plasma VEGF levels. Sunitinib 12-21 vascular endothelial growth factor A Homo sapiens 77-81 23975598-6 2014 In addition, we found that both SK1 and ERK were activated in Sunitinib-resistance cell lines and SK1 and ERK inhibitors could resensitize Sunitinib-resistant cell lines. Sunitinib 139-148 EPH receptor B2 Homo sapiens 40-43 24414551-2 2014 Acute withdrawal of sunitinib results in a proliferative withdrawal flare, primarily due to elevated VEGF levels. Sunitinib 20-29 vascular endothelial growth factor A Homo sapiens 101-105 24414551-14 2014 As predicted, VEGF levels increased during sunitinib exposure followed by a rapid decline after bevacizumab. Sunitinib 43-52 vascular endothelial growth factor A Homo sapiens 14-18 24414551-16 2014 The increase in VEGF at D42 was unexpected based on sunitinib alone and contrary to the hypothesis that we would block VEGF flare with low-dose bevacizumab. Sunitinib 52-61 vascular endothelial growth factor A Homo sapiens 16-20 23975598-6 2014 In addition, we found that both SK1 and ERK were activated in Sunitinib-resistance cell lines and SK1 and ERK inhibitors could resensitize Sunitinib-resistant cell lines. Sunitinib 139-148 sphingosine kinase 1 Homo sapiens 98-101 23975598-6 2014 In addition, we found that both SK1 and ERK were activated in Sunitinib-resistance cell lines and SK1 and ERK inhibitors could resensitize Sunitinib-resistant cell lines. Sunitinib 139-148 EPH receptor B2 Homo sapiens 106-109 23975598-7 2014 In conclusion, our observations suggest that SK1 and ERK activation is a feature of resistant cell lines, which serves as an alternative pathway evading anti-tumor activity of Sunitinib. Sunitinib 176-185 sphingosine kinase 1 Homo sapiens 45-48 23975598-7 2014 In conclusion, our observations suggest that SK1 and ERK activation is a feature of resistant cell lines, which serves as an alternative pathway evading anti-tumor activity of Sunitinib. Sunitinib 176-185 EPH receptor B2 Homo sapiens 53-56 24234588-0 2014 Predictive value of CYP3A and ABCB1 phenotyping probes for the pharmacokinetics of sunitinib: the ClearSun study. Sunitinib 83-92 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 20-25 24234588-0 2014 Predictive value of CYP3A and ABCB1 phenotyping probes for the pharmacokinetics of sunitinib: the ClearSun study. Sunitinib 83-92 ATP binding cassette subfamily B member 1 Homo sapiens 30-35 24393200-7 2014 In vitro NOS enzyme activity assay was used to determine the direct inhibition of nNOS by sunitinib. Sunitinib 90-99 nitric oxide synthase 1 Homo sapiens 82-86 24234588-2 2014 In this prospective study cytochrome P450 (CYP) 3A and adenosine triphosphate binding cassette (ABC) B1 phenotypes were correlated to the pharmacokinetics of sunitinib and its active metabolite N-desethylsunitinib. Sunitinib 158-167 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 26-50 24393200-10 2014 Sunitinib reversed the increase in NO levels, NOS activity, and nNOS expression induced by low potassium or MPP(+) . Sunitinib 0-9 nitric oxide synthase 1 Homo sapiens 64-68 24234588-2 2014 In this prospective study cytochrome P450 (CYP) 3A and adenosine triphosphate binding cassette (ABC) B1 phenotypes were correlated to the pharmacokinetics of sunitinib and its active metabolite N-desethylsunitinib. Sunitinib 158-167 ATP binding cassette subfamily B member 6 (Langereis blood group) Homo sapiens 55-94 24393200-11 2014 Knockdown of nNOS expression partially abolished the neuroprotective effects of sunitinib. Sunitinib 80-89 nitric oxide synthase 1 Homo sapiens 13-17 24234588-2 2014 In this prospective study cytochrome P450 (CYP) 3A and adenosine triphosphate binding cassette (ABC) B1 phenotypes were correlated to the pharmacokinetics of sunitinib and its active metabolite N-desethylsunitinib. Sunitinib 158-167 ATP binding cassette subfamily B member 6 (Langereis blood group) Homo sapiens 96-99 24393200-12 2014 Moreover, sunitinib directly inhibited nNOS enzyme activity. Sunitinib 10-19 nitric oxide synthase 1 Homo sapiens 39-43 24025975-1 2014 Sunitinib is a multiple tyrosine kinase inhibitor of the vascular endothelial growth factor and platelet-derived growth factor pathway and inhibits angiogenesis, cell proliferation, and tumor cell invasion, and stimulates apoptosis. Sunitinib 0-9 vascular endothelial growth factor A Homo sapiens 57-91 24393200-13 2014 CONCLUSIONS: Sunitinib exerts its neuroprotective effects by inhibiting NO overproduction, possibly via the inhibition of nNOS activity and the decrease in nNOS expression. Sunitinib 13-22 nitric oxide synthase 1 Homo sapiens 122-126 24393200-13 2014 CONCLUSIONS: Sunitinib exerts its neuroprotective effects by inhibiting NO overproduction, possibly via the inhibition of nNOS activity and the decrease in nNOS expression. Sunitinib 13-22 nitric oxide synthase 1 Homo sapiens 156-160 24277456-8 2014 Moreover, CDDP-resistant cells were more sensitive to incubation with PDGFRbeta inhibitors such as pazopanib or sunitinib than sensitive cells, a finding consistent with these cells being dependent on this signaling pathway. Sunitinib 112-121 platelet derived growth factor receptor beta Homo sapiens 70-79 24743196-3 2014 Sorafenib, sunitinib, axitinib, and pazopanib are kinase inhibitors that block vascular endothelial growth factor receptor(VEGFR). Sunitinib 11-20 kinase insert domain receptor Homo sapiens 123-128 24120473-11 2014 Sunitinib inhibited VEGF signaling in neoplastic cells and reduced weight and volume of xenograft tumors in mice. Sunitinib 0-9 vascular endothelial growth factor A Mus musculus 20-24 24120473-14 2014 Strategies to reduce autocrine VEGF signaling (eg, with sunitinib) might be used to prevent or treat cancer in patients with Barrett"s esophagus. Sunitinib 56-65 vascular endothelial growth factor A Homo sapiens 31-35 23421954-13 2014 mRCC patients with the CC-genotype in VEGFR1 SNP rs9582036 have a poorer response rate, PFS and OS when treated with sunitinib. Sunitinib 117-126 fms related receptor tyrosine kinase 1 Homo sapiens 38-44 24276455-6 2014 RESULTS: Sunitinib and cisplatin synergistically inhibited the growth of MZ-CRC-1 cells harboring the RET M918T activating mutation. Sunitinib 9-18 ret proto-oncogene Homo sapiens 102-105 24276455-10 2014 CONCLUSIONS: Addition of cisplatin to sunitinib potentiates apoptotic cell death and has promising preclinical activity in MTCs harboring the RET M918T oncogene. Sunitinib 38-47 ret proto-oncogene Homo sapiens 142-145 24575017-6 2014 This report also documents the use of a novel drug combination based on sunitinib that was well tolerated and may warrant testing in other c-kit-dependent cancers. Sunitinib 72-81 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 139-144 23421954-0 2014 VEGFR1 single nucleotide polymorphisms associated with outcome in patients with metastatic renal cell carcinoma treated with sunitinib - a multicentric retrospective analysis. Sunitinib 125-134 fms related receptor tyrosine kinase 1 Homo sapiens 0-6 24475095-4 2014 In vitro experiments were conducted to study the role of miR-942 in sunitinib resistance. Sunitinib 68-77 microRNA 942 Homo sapiens 57-64 24475095-7 2014 MiR-942 was the most accurate predictor of sunitinib efficacy (p = 0.0074). Sunitinib 43-52 microRNA 942 Homo sapiens 0-7 24475095-10 2014 MiR-942 overexpression in Caki-2 up-regulates MMP-9 and VEGF secretion which, in turn, promote HBMEC endothelial migration and sunitinib resistance. Sunitinib 127-136 microRNA 942 Homo sapiens 0-7 24475095-10 2014 MiR-942 overexpression in Caki-2 up-regulates MMP-9 and VEGF secretion which, in turn, promote HBMEC endothelial migration and sunitinib resistance. Sunitinib 127-136 matrix metallopeptidase 9 Homo sapiens 46-51 24475095-10 2014 MiR-942 overexpression in Caki-2 up-regulates MMP-9 and VEGF secretion which, in turn, promote HBMEC endothelial migration and sunitinib resistance. Sunitinib 127-136 vascular endothelial growth factor A Homo sapiens 56-60 24475095-13 2014 We describe a novel paracrine mechanism through which high miR-942 levels in MRCC cells up-regulates MMP-9 and VEGF secretion to enhance endothelial migration and sunitinib resistance. Sunitinib 163-172 microRNA 942 Homo sapiens 59-66 24475095-13 2014 We describe a novel paracrine mechanism through which high miR-942 levels in MRCC cells up-regulates MMP-9 and VEGF secretion to enhance endothelial migration and sunitinib resistance. Sunitinib 163-172 matrix metallopeptidase 9 Homo sapiens 101-106 24475095-13 2014 We describe a novel paracrine mechanism through which high miR-942 levels in MRCC cells up-regulates MMP-9 and VEGF secretion to enhance endothelial migration and sunitinib resistance. Sunitinib 163-172 vascular endothelial growth factor A Homo sapiens 111-115 24967411-13 2014 CONCLUSION: The combination of sunitinib and telmisartan revealed an enhancement of the blockage of the VEGF pathway on renal tumor resulting in a decrease in neoangiogenesis and an increase in necrosis. Sunitinib 31-40 vascular endothelial growth factor A Mus musculus 104-108 24783204-1 2014 In the past few years, therapies targeted at the von Hippel-Lindau (VHL) and hypoxia-inducible factor (HIF) pathways, such as sunitinib and sorafenib, have been developed to treat clear cell renal cell carcinoma (ccRCC). Sunitinib 126-135 von Hippel-Lindau tumor suppressor Homo sapiens 49-66 24149944-2 2014 Since sunitinib is metabolized by cytochrome P450(CYP)3A4, variability in the activity of this enzyme may explain a considerable proportion of this inter-patient variability. Sunitinib 6-15 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 34-57 24220935-6 2014 In multivariate analysis, baseline sVEGFR-3 and IL-8 remained independent predictors of OS in the sunitinib arm, while no independent predictors of outcome remained in the IFN-alpha arm. Sunitinib 98-107 C-X-C motif chemokine ligand 8 Homo sapiens 48-52 24205792-4 2014 Although sunitinib malate, a multi-kinase inhibitor, has shown effectiveness against imatinib-resistant GISTs, recent studies have indicated that some imatinib-resistant GISTs harboring secondary mutations in the KIT activation loop were also resistant to sunitinib. Sunitinib 9-25 KIT proto-oncogene receptor tyrosine kinase Mus musculus 213-216 24205792-4 2014 Although sunitinib malate, a multi-kinase inhibitor, has shown effectiveness against imatinib-resistant GISTs, recent studies have indicated that some imatinib-resistant GISTs harboring secondary mutations in the KIT activation loop were also resistant to sunitinib. Sunitinib 9-18 KIT proto-oncogene receptor tyrosine kinase Mus musculus 213-216 24018642-6 2014 Combination of rapamycin with sunitinib resulted in enhanced cell cycle arrest in G1 phase, which was accompanied with enhanced suppression of mTOR signaling and disruption of the negative feedback loop that activate AKT upon mTORC1 inhibition. Sunitinib 30-39 mechanistic target of rapamycin kinase Homo sapiens 143-147 24018642-6 2014 Combination of rapamycin with sunitinib resulted in enhanced cell cycle arrest in G1 phase, which was accompanied with enhanced suppression of mTOR signaling and disruption of the negative feedback loop that activate AKT upon mTORC1 inhibition. Sunitinib 30-39 AKT serine/threonine kinase 1 Homo sapiens 217-220 24018642-6 2014 Combination of rapamycin with sunitinib resulted in enhanced cell cycle arrest in G1 phase, which was accompanied with enhanced suppression of mTOR signaling and disruption of the negative feedback loop that activate AKT upon mTORC1 inhibition. Sunitinib 30-39 CREB regulated transcription coactivator 1 Mus musculus 226-232 24018642-7 2014 Furthermore, sunitinib and rapamycin displayed synergistic activity against tube formation by human microvessel endothelial cells as well as outgrowth of endothelial tubes and microvessels both in vitro and in vivo, which is associated with down-regulation of VEGF secretion and HIF1alpha expression. Sunitinib 13-22 vascular endothelial growth factor A Homo sapiens 260-264 24018642-7 2014 Furthermore, sunitinib and rapamycin displayed synergistic activity against tube formation by human microvessel endothelial cells as well as outgrowth of endothelial tubes and microvessels both in vitro and in vivo, which is associated with down-regulation of VEGF secretion and HIF1alpha expression. Sunitinib 13-22 hypoxia inducible factor 1 subunit alpha Homo sapiens 279-288 25018885-3 2014 Autoimmune and neurological side effects have been linked to sunitinib"s inhibition of VEGF receptors with a corresponding increase in VEGF levels, which is associated with development of different neuropathies. Sunitinib 61-70 vascular endothelial growth factor A Homo sapiens 87-91 25018885-3 2014 Autoimmune and neurological side effects have been linked to sunitinib"s inhibition of VEGF receptors with a corresponding increase in VEGF levels, which is associated with development of different neuropathies. Sunitinib 61-70 vascular endothelial growth factor A Homo sapiens 135-139 24311637-13 2014 High endothelial c-KIT expression may define a subgroup of patients who will benefit from sunitinib treatment by achieving prolonged PFS. Sunitinib 90-99 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 17-22 24384849-5 2014 Treatment with tyrosine kinase inhibitors (TKIs) such as imatinib, sunitinib, and regorafenib is effective in controlling unresectable disease; however, drug resistance caused by secondary KIT or PDGFRA mutations eventually develops in 90% of cases. Sunitinib 67-76 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 189-192 24384849-5 2014 Treatment with tyrosine kinase inhibitors (TKIs) such as imatinib, sunitinib, and regorafenib is effective in controlling unresectable disease; however, drug resistance caused by secondary KIT or PDGFRA mutations eventually develops in 90% of cases. Sunitinib 67-76 platelet derived growth factor receptor alpha Homo sapiens 196-202 26168132-0 2014 HIF-1 Dimerization Inhibitor Acriflavine Enhances Antitumor Activity of Sunitinib in Breast Cancer Model. Sunitinib 72-81 hypoxia inducible factor 1 subunit alpha Homo sapiens 0-5 24685916-3 2014 She was successfully treated with daily administration of sunitinib malate, an oral multi-target tyrosine kinase inhibitor, which particularly targets vascular endothelial growth factor (VEGF) receptors. Sunitinib 58-74 vascular endothelial growth factor A Homo sapiens 151-185 24685916-3 2014 She was successfully treated with daily administration of sunitinib malate, an oral multi-target tyrosine kinase inhibitor, which particularly targets vascular endothelial growth factor (VEGF) receptors. Sunitinib 58-74 vascular endothelial growth factor A Homo sapiens 187-191 26168132-4 2014 The combination of sunitinib with acriflavine significantly decreased vascular endothelial growth factor and TGF-beta expression and reduced tumor vasculature followed by increased intratumor necrosis and apoptosis. Sunitinib 19-28 vascular endothelial growth factor A Homo sapiens 70-104 26168132-4 2014 The combination of sunitinib with acriflavine significantly decreased vascular endothelial growth factor and TGF-beta expression and reduced tumor vasculature followed by increased intratumor necrosis and apoptosis. Sunitinib 19-28 transforming growth factor beta 1 Homo sapiens 109-117 24756791-1 2014 Sunitinib is an oral multikinase inhibitor that blocks the vascular endothelial growth factor receptor (VEGFR), platelet-derived growth factor receptor (PDGFR) alpha and beta, c-kit, and other receptors. Sunitinib 0-9 kinase insert domain receptor Homo sapiens 59-102 24756791-1 2014 Sunitinib is an oral multikinase inhibitor that blocks the vascular endothelial growth factor receptor (VEGFR), platelet-derived growth factor receptor (PDGFR) alpha and beta, c-kit, and other receptors. Sunitinib 0-9 kinase insert domain receptor Homo sapiens 104-109 24756791-1 2014 Sunitinib is an oral multikinase inhibitor that blocks the vascular endothelial growth factor receptor (VEGFR), platelet-derived growth factor receptor (PDGFR) alpha and beta, c-kit, and other receptors. Sunitinib 0-9 platelet derived growth factor receptor alpha Homo sapiens 153-165 24756791-1 2014 Sunitinib is an oral multikinase inhibitor that blocks the vascular endothelial growth factor receptor (VEGFR), platelet-derived growth factor receptor (PDGFR) alpha and beta, c-kit, and other receptors. Sunitinib 0-9 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 176-181 24324073-6 2013 AIM: This study sought to evaluate the expression of PDGFR alpha/beta and hypoxia-inducible factor-1alpha (HIF-1alpha) and their alterations induced by everolimus, sorafenib and sunitinib in chemonaive HPV-positive and HPV-negative HNSCC. Sunitinib 178-187 platelet derived growth factor receptor alpha Homo sapiens 53-64 25764782-7 2014 Systemic therapies regarding malignant insulinoma are: chemotherapy, somatostatin analogs, radiolabelled somatostatin analogs and the newly developed biological targeted therapies such as everolimus--oral inhibitor of the mammalian target of rapamycin, and sunitinib--the tyrosine kinase inhibitor. Sunitinib 257-266 mechanistic target of rapamycin kinase Homo sapiens 222-251 23848205-0 2013 Curcumin potentiates the ability of sunitinib to eliminate the VHL-lacking renal cancer cells 786-O: rapid inhibition of Rb phosphorylation as a preamble to cyclin D1 inhibition. Sunitinib 36-45 cyclin D1 Homo sapiens 157-166 23848205-9 2013 Since curcumin is known to inhibit the cyclin D1-dependent G1/S-phase kinase CDK4 and the cyclin B-dependent G2/M-phase kinase CDK1 that catalyze phosphorylation-mediated inactivation of Rb, our results indicate that SunC containing a lower dose of sunitinib would be effective in restoring the tumor suppressor activity of Rb, thereby truncating cell cycle and triggering cell death. Sunitinib 249-258 cyclin dependent kinase 1 Homo sapiens 127-131 24324073-6 2013 AIM: This study sought to evaluate the expression of PDGFR alpha/beta and hypoxia-inducible factor-1alpha (HIF-1alpha) and their alterations induced by everolimus, sorafenib and sunitinib in chemonaive HPV-positive and HPV-negative HNSCC. Sunitinib 178-187 hypoxia inducible factor 1 subunit alpha Homo sapiens 107-117 24324073-11 2013 Incubation with everolimus, sorafenib or sunitinib led to a decrease in PDGFR alpha/beta and HIF-1alpha expression, depending on the HPV status. Sunitinib 41-50 platelet derived growth factor receptor alpha Homo sapiens 72-83 24324073-11 2013 Incubation with everolimus, sorafenib or sunitinib led to a decrease in PDGFR alpha/beta and HIF-1alpha expression, depending on the HPV status. Sunitinib 41-50 hypoxia inducible factor 1 subunit alpha Homo sapiens 93-103 24295410-0 2013 Sunitinib in malignant melanoma: a treatment option only for KIT-mutated patients? Sunitinib 0-9 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 61-64 24061866-6 2013 Using specific human and mouse probes for genes encoding sunitinib targets, we showed a significant relation between apoptotic tumor cell numbers and human PDGFRBeta and RET mRNA expression in liver cancer and to human VEGFR2 expression in breast cancer xenografts. Sunitinib 57-66 platelet derived growth factor receptor beta Homo sapiens 156-165 24061866-6 2013 Using specific human and mouse probes for genes encoding sunitinib targets, we showed a significant relation between apoptotic tumor cell numbers and human PDGFRBeta and RET mRNA expression in liver cancer and to human VEGFR2 expression in breast cancer xenografts. Sunitinib 57-66 ret proto-oncogene Homo sapiens 170-173 24061866-6 2013 Using specific human and mouse probes for genes encoding sunitinib targets, we showed a significant relation between apoptotic tumor cell numbers and human PDGFRBeta and RET mRNA expression in liver cancer and to human VEGFR2 expression in breast cancer xenografts. Sunitinib 57-66 kinase insert domain receptor Homo sapiens 219-225 24122794-0 2013 Autotaxin-lysophosphatidic acid signaling axis mediates tumorigenesis and development of acquired resistance to sunitinib in renal cell carcinoma. Sunitinib 112-121 ectonucleotide pyrophosphatase/phosphodiesterase 2 Homo sapiens 0-9 24122794-5 2013 RESULTS: Altered expression of autotaxin, an extracellular lysophospholipase D, was detected in sunitinib-treated tumor vasculature of human RCC and in the tumor endothelial cells of RCC xenograft models when adapting to sunitinib. Sunitinib 96-105 ectonucleotide pyrophosphatase/phosphodiesterase 2 Homo sapiens 31-40 24122794-5 2013 RESULTS: Altered expression of autotaxin, an extracellular lysophospholipase D, was detected in sunitinib-treated tumor vasculature of human RCC and in the tumor endothelial cells of RCC xenograft models when adapting to sunitinib. Sunitinib 96-105 ectonucleotide pyrophosphatase/phosphodiesterase 2 Homo sapiens 45-78 24122794-5 2013 RESULTS: Altered expression of autotaxin, an extracellular lysophospholipase D, was detected in sunitinib-treated tumor vasculature of human RCC and in the tumor endothelial cells of RCC xenograft models when adapting to sunitinib. Sunitinib 221-230 ectonucleotide pyrophosphatase/phosphodiesterase 2 Homo sapiens 31-40 24122794-5 2013 RESULTS: Altered expression of autotaxin, an extracellular lysophospholipase D, was detected in sunitinib-treated tumor vasculature of human RCC and in the tumor endothelial cells of RCC xenograft models when adapting to sunitinib. Sunitinib 221-230 ectonucleotide pyrophosphatase/phosphodiesterase 2 Homo sapiens 45-78 24122794-10 2013 In addition, coadministration of Ki16425 with sunitinib prolonged the sensitivity of RCC to sunitinib in xenograft models, suggesting that ATX-LPA signaling in part mediates the acquired resistance against sunitinib in RCC. Sunitinib 46-55 ectonucleotide pyrophosphatase/phosphodiesterase 2 Homo sapiens 139-142 24122794-10 2013 In addition, coadministration of Ki16425 with sunitinib prolonged the sensitivity of RCC to sunitinib in xenograft models, suggesting that ATX-LPA signaling in part mediates the acquired resistance against sunitinib in RCC. Sunitinib 92-101 ectonucleotide pyrophosphatase/phosphodiesterase 2 Homo sapiens 139-142 24122794-10 2013 In addition, coadministration of Ki16425 with sunitinib prolonged the sensitivity of RCC to sunitinib in xenograft models, suggesting that ATX-LPA signaling in part mediates the acquired resistance against sunitinib in RCC. Sunitinib 92-101 ectonucleotide pyrophosphatase/phosphodiesterase 2 Homo sapiens 139-142 24122794-11 2013 CONCLUSIONS: Our results reveal that endothelial ATX acts through LPA signaling to promote renal tumorigenesis and is functionally involved in the acquired resistance of RCC to sunitinib. Sunitinib 177-186 ectonucleotide pyrophosphatase/phosphodiesterase 2 Homo sapiens 49-52 24295410-1 2013 Sunitinib has previously been reported to be potentially effective in the treatment of malignant melanomas expressing c-KIT. Sunitinib 0-9 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 118-123 24295410-4 2013 To our knowledge, this represents the first evidence of sunitinib activity in KIT wild-type melanoma. Sunitinib 56-65 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 78-81 23812726-7 2013 The inhibition of kinase activation using multi-targeted kinase inhibitors, sorafenib and sunitinib led to significant cell growth inhibition and apoptosis induction via suppression of Erk1/2 and Akt activation, whereas drugs with specificity to a single kinase showed less potency. Sunitinib 90-99 mitogen-activated protein kinase 3 Homo sapiens 185-191 24113148-5 2013 Furthermore, the distribution of sunitinib to the brain increased after administration of selective P-glycoprotein (P-gp) or breast cancer resistance protein (Bcrp) pharmacological inhibitors and a dual inhibitor, elacridar, comparable to that of the corresponding transgenic genotype. Sunitinib 33-42 phosphoglycolate phosphatase Mus musculus 100-114 24113148-5 2013 Furthermore, the distribution of sunitinib to the brain increased after administration of selective P-glycoprotein (P-gp) or breast cancer resistance protein (Bcrp) pharmacological inhibitors and a dual inhibitor, elacridar, comparable to that of the corresponding transgenic genotype. Sunitinib 33-42 phosphoglycolate phosphatase Mus musculus 116-120 24113148-5 2013 Furthermore, the distribution of sunitinib to the brain increased after administration of selective P-glycoprotein (P-gp) or breast cancer resistance protein (Bcrp) pharmacological inhibitors and a dual inhibitor, elacridar, comparable to that of the corresponding transgenic genotype. Sunitinib 33-42 ATP binding cassette subfamily G member 2 (Junior blood group) Mus musculus 125-157 24113148-5 2013 Furthermore, the distribution of sunitinib to the brain increased after administration of selective P-glycoprotein (P-gp) or breast cancer resistance protein (Bcrp) pharmacological inhibitors and a dual inhibitor, elacridar, comparable to that of the corresponding transgenic genotype. Sunitinib 33-42 ATP binding cassette subfamily G member 2 (Junior blood group) Mus musculus 159-163 24167056-5 2013 Among the differentially expressed proteins, heme oxygenase-1 (HO-1) was found in the sunitinib and imatinib mesylate treatment groups, which possibly acts as a specific target for synthetic lethality in combinational treatment. Sunitinib 86-95 heme oxygenase 1 Homo sapiens 45-61 24167056-5 2013 Among the differentially expressed proteins, heme oxygenase-1 (HO-1) was found in the sunitinib and imatinib mesylate treatment groups, which possibly acts as a specific target for synthetic lethality in combinational treatment. Sunitinib 86-95 heme oxygenase 1 Homo sapiens 63-67 23812726-7 2013 The inhibition of kinase activation using multi-targeted kinase inhibitors, sorafenib and sunitinib led to significant cell growth inhibition and apoptosis induction via suppression of Erk1/2 and Akt activation, whereas drugs with specificity to a single kinase showed less potency. Sunitinib 90-99 AKT serine/threonine kinase 1 Homo sapiens 196-199 24222145-0 2013 VEGF expression and response to sunitinib in patients with metastatic clear cell renal cell carcinoma. Sunitinib 32-41 vascular endothelial growth factor A Homo sapiens 0-4 24107802-0 2013 Hyperbilirubinemia in pazopanib- or sunitinib-treated patients in COMPARZ is associated with UGT1A1 polymorphisms. Sunitinib 36-45 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-99 24257372-0 2013 PKPD Modeling of VEGF, sVEGFR-2, sVEGFR-3, and sKIT as Predictors of Tumor Dynamics and Overall Survival Following Sunitinib Treatment in GIST. Sunitinib 115-124 vascular endothelial growth factor A Homo sapiens 17-21 24232460-0 2013 Sunitinib indirectly enhanced anti-tumor cytotoxicity of cytokine-induced killer cells and CD3+CD56+ subset through the co-culturing dendritic cells. Sunitinib 0-9 neural cell adhesion molecule 1 Homo sapiens 95-99 24232460-5 2013 Sunitinib promoted Th1-inducing and pro-inflammatory phenotypes (IL-12, IFN-gamma and IL-6) in DCs at the expense of Th2 inducing phenotype (IL-13) and regulatory phenotype (PD-L1, IDO). Sunitinib 0-9 negative elongation factor complex member C/D Homo sapiens 19-22 24232460-5 2013 Sunitinib promoted Th1-inducing and pro-inflammatory phenotypes (IL-12, IFN-gamma and IL-6) in DCs at the expense of Th2 inducing phenotype (IL-13) and regulatory phenotype (PD-L1, IDO). Sunitinib 0-9 interferon gamma Homo sapiens 72-81 24232460-5 2013 Sunitinib promoted Th1-inducing and pro-inflammatory phenotypes (IL-12, IFN-gamma and IL-6) in DCs at the expense of Th2 inducing phenotype (IL-13) and regulatory phenotype (PD-L1, IDO). Sunitinib 0-9 interleukin 6 Homo sapiens 86-90 24232460-5 2013 Sunitinib promoted Th1-inducing and pro-inflammatory phenotypes (IL-12, IFN-gamma and IL-6) in DCs at the expense of Th2 inducing phenotype (IL-13) and regulatory phenotype (PD-L1, IDO). Sunitinib 0-9 CD274 molecule Homo sapiens 174-179 24232460-5 2013 Sunitinib promoted Th1-inducing and pro-inflammatory phenotypes (IL-12, IFN-gamma and IL-6) in DCs at the expense of Th2 inducing phenotype (IL-13) and regulatory phenotype (PD-L1, IDO). Sunitinib 0-9 indoleamine 2,3-dioxygenase 1 Homo sapiens 181-184 24232460-6 2013 Sunitinib-treated DCs subsequently induced the upregulation of Th1 phenotypic markers (IFN-gamma and T-bet) and the downregulation of the Th2 signature (GATA-3) and the Th17 marker (RORC) on the CD3+CD56+ subset of CIK cells. Sunitinib 0-9 negative elongation factor complex member C/D Homo sapiens 63-66 24232460-6 2013 Sunitinib-treated DCs subsequently induced the upregulation of Th1 phenotypic markers (IFN-gamma and T-bet) and the downregulation of the Th2 signature (GATA-3) and the Th17 marker (RORC) on the CD3+CD56+ subset of CIK cells. Sunitinib 0-9 interferon gamma Homo sapiens 87-96 24232460-6 2013 Sunitinib-treated DCs subsequently induced the upregulation of Th1 phenotypic markers (IFN-gamma and T-bet) and the downregulation of the Th2 signature (GATA-3) and the Th17 marker (RORC) on the CD3+CD56+ subset of CIK cells. Sunitinib 0-9 T-box transcription factor 21 Homo sapiens 101-106 24232460-6 2013 Sunitinib-treated DCs subsequently induced the upregulation of Th1 phenotypic markers (IFN-gamma and T-bet) and the downregulation of the Th2 signature (GATA-3) and the Th17 marker (RORC) on the CD3+CD56+ subset of CIK cells. Sunitinib 0-9 GATA binding protein 3 Homo sapiens 153-159 24232460-6 2013 Sunitinib-treated DCs subsequently induced the upregulation of Th1 phenotypic markers (IFN-gamma and T-bet) and the downregulation of the Th2 signature (GATA-3) and the Th17 marker (RORC) on the CD3+CD56+ subset of CIK cells. Sunitinib 0-9 neural cell adhesion molecule 1 Homo sapiens 199-203 24232460-7 2013 It concluded that sunitinib-pretreated DCs drove the CD3+CD56+ subset toward Th1 phenotype with increased anti-tumor cytotoxicity. Sunitinib 18-27 neural cell adhesion molecule 1 Homo sapiens 57-61 24232460-7 2013 It concluded that sunitinib-pretreated DCs drove the CD3+CD56+ subset toward Th1 phenotype with increased anti-tumor cytotoxicity. Sunitinib 18-27 negative elongation factor complex member C/D Homo sapiens 77-80 24223213-10 2013 Furthermore, treatment with two anti-proliferative, multi-receptor tyrosine kinase inhibitors-sunitinib and sorafenib-strongly upregulated biglycan expression. Sunitinib 94-103 biglycan Homo sapiens 139-147 24222145-5 2013 Patients with higher H-score and higher VEGF expression had a significantly shorter survival; OS after first-line sunitinib therapy and PFS correlated with MSKCC score and DMFS but not with VEGF expression and H score. Sunitinib 114-123 vascular endothelial growth factor A Homo sapiens 40-44 24222145-1 2013 AIM: To verify whether vascular endothelial growth factor (VEGF) is associated with distant metastasis free survival (DMFS) and Overall Survival (OS) of patients with renal cell carcinoma (RCC) treated with sunitinib. Sunitinib 207-216 vascular endothelial growth factor A Homo sapiens 23-57 24222145-1 2013 AIM: To verify whether vascular endothelial growth factor (VEGF) is associated with distant metastasis free survival (DMFS) and Overall Survival (OS) of patients with renal cell carcinoma (RCC) treated with sunitinib. Sunitinib 207-216 vascular endothelial growth factor A Homo sapiens 59-63 23969938-0 2013 Emergence of polyclonal FLT3 tyrosine kinase domain mutations during sequential therapy with sorafenib and sunitinib in FLT3-ITD-positive acute myeloid leukemia. Sunitinib 107-116 fms related receptor tyrosine kinase 3 Homo sapiens 24-28 23471661-0 2013 Sunitinib reverse multidrug resistance in gastric cancer cells by modulating Stat3 and inhibiting P-gp function. Sunitinib 0-9 signal transducer and activator of transcription 3 Homo sapiens 77-82 23471661-0 2013 Sunitinib reverse multidrug resistance in gastric cancer cells by modulating Stat3 and inhibiting P-gp function. Sunitinib 0-9 phosphoglycolate phosphatase Homo sapiens 98-102 23471661-6 2013 In addition, the present study revealed that Sunitinib inhibited Stat3 and down-regulated Bcl-2 in SGC7901/VCR cells, which might also contribute to the reversal of MDR. Sunitinib 45-54 signal transducer and activator of transcription 3 Homo sapiens 65-70 23471661-6 2013 In addition, the present study revealed that Sunitinib inhibited Stat3 and down-regulated Bcl-2 in SGC7901/VCR cells, which might also contribute to the reversal of MDR. Sunitinib 45-54 BCL2 apoptosis regulator Homo sapiens 90-95 23471661-7 2013 In conclusion, Sunitinib reverses multidrug resistance in gastric cancer cells by inhibiting P-gp transporter function and modulating Stat3 and Bcl-2. Sunitinib 15-24 phosphoglycolate phosphatase Homo sapiens 93-97 23471661-7 2013 In conclusion, Sunitinib reverses multidrug resistance in gastric cancer cells by inhibiting P-gp transporter function and modulating Stat3 and Bcl-2. Sunitinib 15-24 signal transducer and activator of transcription 3 Homo sapiens 134-139 23471661-7 2013 In conclusion, Sunitinib reverses multidrug resistance in gastric cancer cells by inhibiting P-gp transporter function and modulating Stat3 and Bcl-2. Sunitinib 15-24 BCL2 apoptosis regulator Homo sapiens 144-149 23659419-0 2013 Acute renal failure during the "off" period after sunitinib administration: possible mechanism of vascular endothelial growth factor cascade hyperactivation. Sunitinib 50-59 vascular endothelial growth factor A Homo sapiens 98-132 24204982-8 2013 We determined the 1.4 A co-crystal structure of PAK6 with the type II PAK inhibitor PF-3758309, and the 1.95 A co-crystal structure of PAK6 with sunitinib. Sunitinib 145-154 p21 (RAC1) activated kinase 6 Mus musculus 135-139 23969938-1 2013 PURPOSE: To evaluate the clinical activity of sequential therapy with sorafenib and sunitinib in FMS-like tyrosine kinase 3 (FLT3)-internal tandem duplication (ITD)-positive acute myelogenous leukemia (AML) and monitor the emergence of secondary FLT3 tyrosine kinase domain (TKD) mutations during treatment. Sunitinib 84-93 fms related receptor tyrosine kinase 3 Homo sapiens 97-123 23969938-1 2013 PURPOSE: To evaluate the clinical activity of sequential therapy with sorafenib and sunitinib in FMS-like tyrosine kinase 3 (FLT3)-internal tandem duplication (ITD)-positive acute myelogenous leukemia (AML) and monitor the emergence of secondary FLT3 tyrosine kinase domain (TKD) mutations during treatment. Sunitinib 84-93 fms related receptor tyrosine kinase 3 Homo sapiens 125-129 23969938-13 2013 Cells expressing sorafenib-resistant FLT3 mutations were sensitive to sunitinib in vitro. Sunitinib 70-79 fms related receptor tyrosine kinase 3 Homo sapiens 37-41 23969938-14 2013 CONCLUSIONS: Sunitinib has activity in patients that are resistant to sorafenib and harbor secondary FLT3 TKD mutations. Sunitinib 13-22 fms related receptor tyrosine kinase 3 Homo sapiens 101-105 23716000-6 2013 Supplement with recombinant mouse IL-12b or restoration of IL-12b expression in the host by zoledronic acid, which was previously reported to enhance IL-12 expression in vitro and in vivo, alleviated the metastasis-promoting effects of sunitinib and sorafenib. Sunitinib 236-245 interleukin 12b Mus musculus 34-40 23716000-6 2013 Supplement with recombinant mouse IL-12b or restoration of IL-12b expression in the host by zoledronic acid, which was previously reported to enhance IL-12 expression in vitro and in vivo, alleviated the metastasis-promoting effects of sunitinib and sorafenib. Sunitinib 236-245 interleukin 12b Mus musculus 59-65 24123039-1 2013 BACKGROUND/AIM: We analyzed the efficacy and toxicity profile of sunitinib according to single nucleotide polymorphisms (SNPs) of vascular endothelial growth factor receptor (VEGFR) and Kinase insert domain receptor (KDR). Sunitinib 65-74 kinase insert domain receptor Homo sapiens 130-173 24123039-9 2013 CONCLUSION: Certain SNPs of KDR may affect treatment outcome and toxicity in patients treated with sunitinib. Sunitinib 99-108 kinase insert domain receptor Homo sapiens 28-31 24013576-8 2013 CONCLUSION: Among 12 genetic polymorphisms, polymorphism in the ABCG2 421C>A gene may be mostly associated with the risk of sunitinib-related toxicity in mRCC patients. Sunitinib 127-136 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 64-69 23707608-7 2013 The multi-targeted TKi sunitinib showed decreased cardiomyocyte viability, AMPK inhibition, increased lipid accumulation, disrupted beat pattern, and hERG block. Sunitinib 23-32 ETS transcription factor ERG Homo sapiens 150-154 23878397-0 2013 Platelet-derived growth factor/vascular endothelial growth factor receptor inactivation by sunitinib results in Tsc1/Tsc2-dependent inhibition of TORC1. Sunitinib 91-100 vascular endothelial growth factor A Homo sapiens 31-65 23878397-0 2013 Platelet-derived growth factor/vascular endothelial growth factor receptor inactivation by sunitinib results in Tsc1/Tsc2-dependent inhibition of TORC1. Sunitinib 91-100 TSC complex subunit 1 Homo sapiens 112-116 23878397-0 2013 Platelet-derived growth factor/vascular endothelial growth factor receptor inactivation by sunitinib results in Tsc1/Tsc2-dependent inhibition of TORC1. Sunitinib 91-100 TSC complex subunit 2 Homo sapiens 117-121 23878397-0 2013 Platelet-derived growth factor/vascular endothelial growth factor receptor inactivation by sunitinib results in Tsc1/Tsc2-dependent inhibition of TORC1. Sunitinib 91-100 CREB regulated transcription coactivator 1 Homo sapiens 146-151 23878397-1 2013 Vascular endothelial growth factor (VEGF) and platelet-derived growth factor (PDGF) receptors are implicated in development and tumorigenesis and dual inhibitors like sunitinib are prescribed for cancer treatment. Sunitinib 167-176 vascular endothelial growth factor A Homo sapiens 0-34 23878397-1 2013 Vascular endothelial growth factor (VEGF) and platelet-derived growth factor (PDGF) receptors are implicated in development and tumorigenesis and dual inhibitors like sunitinib are prescribed for cancer treatment. Sunitinib 167-176 vascular endothelial growth factor A Homo sapiens 36-40 23878397-1 2013 Vascular endothelial growth factor (VEGF) and platelet-derived growth factor (PDGF) receptors are implicated in development and tumorigenesis and dual inhibitors like sunitinib are prescribed for cancer treatment. Sunitinib 167-176 PDGF- and VEGF-receptor related Drosophila melanogaster 78-82 23878397-9 2013 Like its mammalian orthologs, PVR is inhibited by sunitinib, and sunitinib treatment phenocopies PVR loss in hemocytes. Sunitinib 50-59 PVR cell adhesion molecule Homo sapiens 30-33 23878397-9 2013 Like its mammalian orthologs, PVR is inhibited by sunitinib, and sunitinib treatment phenocopies PVR loss in hemocytes. Sunitinib 65-74 PVR cell adhesion molecule Homo sapiens 97-100 23878397-10 2013 Sunitinib inhibits TORC1 in insect cells, and sunitinib-mediated TORC1 inhibition requires an intact Tsc1/Tsc2 complex. Sunitinib 0-9 CREB regulated transcription coactivator 1 Homo sapiens 19-24 23878397-10 2013 Sunitinib inhibits TORC1 in insect cells, and sunitinib-mediated TORC1 inhibition requires an intact Tsc1/Tsc2 complex. Sunitinib 46-55 CREB regulated transcription coactivator 1 Homo sapiens 65-70 23878397-10 2013 Sunitinib inhibits TORC1 in insect cells, and sunitinib-mediated TORC1 inhibition requires an intact Tsc1/Tsc2 complex. Sunitinib 46-55 TSC complex subunit 1 Homo sapiens 101-105 23878397-10 2013 Sunitinib inhibits TORC1 in insect cells, and sunitinib-mediated TORC1 inhibition requires an intact Tsc1/Tsc2 complex. Sunitinib 46-55 TSC complex subunit 2 Homo sapiens 106-110 23878397-11 2013 Sunitinib similarly inhibited TORC1 in human endothelial cells in a Tsc1/Tsc2-dependent manner. Sunitinib 0-9 CREB regulated transcription coactivator 1 Homo sapiens 30-35 23878397-11 2013 Sunitinib similarly inhibited TORC1 in human endothelial cells in a Tsc1/Tsc2-dependent manner. Sunitinib 0-9 TSC complex subunit 1 Homo sapiens 68-72 23878397-11 2013 Sunitinib similarly inhibited TORC1 in human endothelial cells in a Tsc1/Tsc2-dependent manner. Sunitinib 0-9 TSC complex subunit 2 Homo sapiens 73-77 23878397-12 2013 Our findings provide insight into the mechanism of action of PVR and may have implications for understanding sunitinib sensitivity and resistance in tumors. Sunitinib 109-118 PVR cell adhesion molecule Homo sapiens 61-64 24082917-2 2013 Currently available oral VEGF tyrosine kinase inhibitors (TKIs) approved for treatment of mRCC include sorafenib, sunitinib, pazopanib, and axitinib. Sunitinib 114-123 vascular endothelial growth factor A Homo sapiens 25-29 24086736-8 2013 RESULTS: Immunostaining of HIF-1alpha, CA9, Ki67, CD31, pVEGFR1, VEGFR1 and -2, pPDGFRalpha and -beta was significantly associated with the sunitinib response after 6 and 9 months as well as last report under therapy. Sunitinib 140-149 hypoxia inducible factor 1 subunit alpha Homo sapiens 27-37 24086736-8 2013 RESULTS: Immunostaining of HIF-1alpha, CA9, Ki67, CD31, pVEGFR1, VEGFR1 and -2, pPDGFRalpha and -beta was significantly associated with the sunitinib response after 6 and 9 months as well as last report under therapy. Sunitinib 140-149 fms related receptor tyrosine kinase 1 Homo sapiens 65-78 23788753-5 2013 RESULTS: High expression of HIF2A and PDGFRB was associated with better sunitinib RECIST objective response (P = 0.024 and P = 0.026; respectively) and increased VEGFR3 expression was associated with longer progression-free survival (P = 0.012). Sunitinib 72-81 endothelial PAS domain protein 1 Homo sapiens 28-33 24073208-0 2013 EMMPRIN promotes angiogenesis, proliferation, invasion and resistance to sunitinib in renal cell carcinoma, and its level predicts patient outcome. Sunitinib 73-82 basigin (Ok blood group) Homo sapiens 0-7 24073208-7 2013 EMMPRIN expression was evaluated in RCC patients who received sunitinib therapy and in sunitinib-resistant cells. Sunitinib 62-71 basigin (Ok blood group) Homo sapiens 0-7 24073208-7 2013 EMMPRIN expression was evaluated in RCC patients who received sunitinib therapy and in sunitinib-resistant cells. Sunitinib 87-96 basigin (Ok blood group) Homo sapiens 0-7 24073208-8 2013 Further, the relation between EMMPRIN expression and sensitivity to sunitinib was examined. Sunitinib 68-77 basigin (Ok blood group) Homo sapiens 30-37 24073208-12 2013 EMMPRIN expression was significantly increased in patients who received sunitinib therapy as well as in sunitinib-resistant 786-O cells (786-suni). Sunitinib 72-81 basigin (Ok blood group) Homo sapiens 0-7 24073208-12 2013 EMMPRIN expression was significantly increased in patients who received sunitinib therapy as well as in sunitinib-resistant 786-O cells (786-suni). Sunitinib 104-113 basigin (Ok blood group) Homo sapiens 0-7 24073208-13 2013 EMMPRIN-overexpressing RCC cells were resistant to sunitinib. Sunitinib 51-60 basigin (Ok blood group) Homo sapiens 0-7 24073208-14 2013 CONCLUSION: Our findings indicate that high expression of EMMPRIN in RCC plays important roles in tumor progression and sunitinib resistance. Sunitinib 120-129 basigin (Ok blood group) Homo sapiens 58-65 23788753-5 2013 RESULTS: High expression of HIF2A and PDGFRB was associated with better sunitinib RECIST objective response (P = 0.024 and P = 0.026; respectively) and increased VEGFR3 expression was associated with longer progression-free survival (P = 0.012). Sunitinib 72-81 platelet derived growth factor receptor beta Homo sapiens 38-44 23788753-6 2013 VEGFR3 overexpression showed a negative correlation with VEGFR3 polymorphism rs307826 (P = 0.002), a sunitinib resistance predictor. Sunitinib 101-110 fms related receptor tyrosine kinase 4 Homo sapiens 0-6 23788753-6 2013 VEGFR3 overexpression showed a negative correlation with VEGFR3 polymorphism rs307826 (P = 0.002), a sunitinib resistance predictor. Sunitinib 101-110 fms related receptor tyrosine kinase 4 Homo sapiens 57-63 23788753-10 2013 In addition, low VEGFR3 expression was associated with worse outcome and with VEGFR3 rs307826 variant allele, reinforcing VEGFR3 as a marker of sunitinib resistance. Sunitinib 144-153 fms related receptor tyrosine kinase 4 Homo sapiens 17-23 25841679-10 2013 Biologic agents targeting the VEGF and mTOR signaling pathways, e.g., sunitinib, bevacizumab or everolimus are becoming integrated as treatments for PNET"s. Sunitinib 70-79 vascular endothelial growth factor A Homo sapiens 30-34 25841679-10 2013 Biologic agents targeting the VEGF and mTOR signaling pathways, e.g., sunitinib, bevacizumab or everolimus are becoming integrated as treatments for PNET"s. Sunitinib 70-79 mechanistic target of rapamycin kinase Homo sapiens 39-43 23475388-8 2013 CONCLUSIONS: Our findings showing constant expression levels of VEGFR2 in endothelial cells serve as a first indication that the use of small tyrosine kinase inhibitors such as Sunitinib directly targeting the VEGF-receptors might be worth testing, also in the clinical context in cases of therapy-refractory meningiomas. Sunitinib 177-186 kinase insert domain receptor Homo sapiens 64-70 23720233-4 2013 Inhibition of phosphoinositide 3-kinase (PI3K) by LY294002 treatment and application of sunitinib, a widely used anti-angiogenic compound, suppressed enhanced angiogenesis in Pten mutants. Sunitinib 88-97 phosphatase and tensin homolog B Danio rerio 175-179 23720233-8 2013 Surprisingly, sunitinib treatment dramatically enhanced vegfaa expression in Pten mutant embryos, which might account for tumor relapse in human patients who are treated with sunitinib. Sunitinib 14-23 vascular endothelial growth factor Aa Danio rerio 56-62 23720233-8 2013 Surprisingly, sunitinib treatment dramatically enhanced vegfaa expression in Pten mutant embryos, which might account for tumor relapse in human patients who are treated with sunitinib. Sunitinib 14-23 phosphatase and tensin homolog Homo sapiens 77-81 23720233-8 2013 Surprisingly, sunitinib treatment dramatically enhanced vegfaa expression in Pten mutant embryos, which might account for tumor relapse in human patients who are treated with sunitinib. Sunitinib 175-184 vascular endothelial growth factor Aa Danio rerio 56-62 23720233-8 2013 Surprisingly, sunitinib treatment dramatically enhanced vegfaa expression in Pten mutant embryos, which might account for tumor relapse in human patients who are treated with sunitinib. Sunitinib 175-184 phosphatase and tensin homolog Homo sapiens 77-81 23720233-9 2013 Combined treatment with suboptimal concentrations of sunitinib and LY294002 rescued enhanced angiogenesis in pten mutant embryos without the dramatic increase in vegfaa expression, suggesting a new approach for therapeutic intervention in VEGFR-signaling-dependent tumors. Sunitinib 53-62 phosphatase and tensin homolog B Danio rerio 109-113 23942298-8 2013 The role of KIT mutation and amplification in the development of ovarian dysgerminoma and the use of Sunitinib, a receptor tyrosine kinase inhibitor with an effect on vascular endothelial growth factor, platelet-derived growth factor and KIT receptors in patients with platinum-resistant GCT, are novel promising approaches. Sunitinib 101-110 vascular endothelial growth factor A Homo sapiens 167-201 23942298-8 2013 The role of KIT mutation and amplification in the development of ovarian dysgerminoma and the use of Sunitinib, a receptor tyrosine kinase inhibitor with an effect on vascular endothelial growth factor, platelet-derived growth factor and KIT receptors in patients with platinum-resistant GCT, are novel promising approaches. Sunitinib 101-110 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 238-241 23767831-2 2013 Prior attempts to block vascular endothelial growth factor (VEGF) with sunitinib, sorafenib and thalidomide have obtained disappointing results. Sunitinib 71-80 vascular endothelial growth factor A Homo sapiens 24-58 23767831-2 2013 Prior attempts to block vascular endothelial growth factor (VEGF) with sunitinib, sorafenib and thalidomide have obtained disappointing results. Sunitinib 71-80 vascular endothelial growth factor A Homo sapiens 60-64 23475388-8 2013 CONCLUSIONS: Our findings showing constant expression levels of VEGFR2 in endothelial cells serve as a first indication that the use of small tyrosine kinase inhibitors such as Sunitinib directly targeting the VEGF-receptors might be worth testing, also in the clinical context in cases of therapy-refractory meningiomas. Sunitinib 177-186 vascular endothelial growth factor A Homo sapiens 64-68 23034406-1 2013 Sorafenib and sunitinib are oral tyrosine kinase inhibitors, commonly used in the treatment of metastatic renal cell carcinoma. Sunitinib 14-23 TXK tyrosine kinase Homo sapiens 33-48 23747315-4 2013 Analysis of cytokine milieu within TME revealed IL-10, TGFbeta, IL-6 rich type 2 characters was significantly switched to type 1 microenvironment with dominance of IFNgamma and IL-2 within NLGP-TME, which was not found in other cases; however Cisplatin-TME appeared better in type 2 to type 1 conversion than Sutent-TME as evidenced by RT-PCR, ELISA and immunohistochemical analysis. Sunitinib 309-315 interleukin 10 Mus musculus 48-53 23747315-4 2013 Analysis of cytokine milieu within TME revealed IL-10, TGFbeta, IL-6 rich type 2 characters was significantly switched to type 1 microenvironment with dominance of IFNgamma and IL-2 within NLGP-TME, which was not found in other cases; however Cisplatin-TME appeared better in type 2 to type 1 conversion than Sutent-TME as evidenced by RT-PCR, ELISA and immunohistochemical analysis. Sunitinib 309-315 interleukin 6 Mus musculus 64-68 23969186-0 2013 Antiproliferative and proapoptotic activity of sunitinib on endothelial and anaplastic thyroid cancer cells via inhibition of Akt and ERK1/2 phosphorylation and by down-regulation of cyclin-D1. Sunitinib 47-56 AKT serine/threonine kinase 1 Homo sapiens 126-129 23969186-0 2013 Antiproliferative and proapoptotic activity of sunitinib on endothelial and anaplastic thyroid cancer cells via inhibition of Akt and ERK1/2 phosphorylation and by down-regulation of cyclin-D1. Sunitinib 47-56 mitogen-activated protein kinase 3 Homo sapiens 134-140 23969186-0 2013 Antiproliferative and proapoptotic activity of sunitinib on endothelial and anaplastic thyroid cancer cells via inhibition of Akt and ERK1/2 phosphorylation and by down-regulation of cyclin-D1. Sunitinib 47-56 cyclin D1 Homo sapiens 183-192 23969186-9 2013 Phospho-vascular endothelial growth factor receptor-2 levels significantly decreased after sunitinib treatment in activated endothelial cells. Sunitinib 91-100 kinase insert domain receptor Homo sapiens 8-53 23969186-10 2013 Phospho-epidermal growth factor receptor, ERK1/2, and Akt phosphorylation was significantly inhibited by sunitinib treatment in endothelial and cancer cells, and cyclin-D1 mRNA and protein expression was inhibited. Sunitinib 105-114 mitogen-activated protein kinase 3 Homo sapiens 42-48 23969186-10 2013 Phospho-epidermal growth factor receptor, ERK1/2, and Akt phosphorylation was significantly inhibited by sunitinib treatment in endothelial and cancer cells, and cyclin-D1 mRNA and protein expression was inhibited. Sunitinib 105-114 AKT serine/threonine kinase 1 Homo sapiens 54-57 23969186-12 2013 CONCLUSIONS: Sunitinib is active in vitro and in vivo against activated endothelial and ATC cells via the inhibition of Akt and ERK1/2 phosphorylation and through the down-regulation of cyclin-D1. Sunitinib 13-22 AKT serine/threonine kinase 1 Homo sapiens 120-123 23969186-12 2013 CONCLUSIONS: Sunitinib is active in vitro and in vivo against activated endothelial and ATC cells via the inhibition of Akt and ERK1/2 phosphorylation and through the down-regulation of cyclin-D1. Sunitinib 13-22 mitogen-activated protein kinase 3 Homo sapiens 128-134 23969186-12 2013 CONCLUSIONS: Sunitinib is active in vitro and in vivo against activated endothelial and ATC cells via the inhibition of Akt and ERK1/2 phosphorylation and through the down-regulation of cyclin-D1. Sunitinib 13-22 cyclin D1 Homo sapiens 186-195 23931237-0 2013 Re: Cox-2 inhibition enhances the activity of sunitinib in human renal cell carcinoma xenografts. Sunitinib 46-55 mitochondrially encoded cytochrome c oxidase II Homo sapiens 4-9 23843632-0 2013 Impact of P-glycoprotein (ABCB1) and breast cancer resistance protein (ABCG2) gene dosage on plasma pharmacokinetics and brain accumulation of dasatinib, sorafenib, and sunitinib. Sunitinib 169-178 ATP binding cassette subfamily G member 2 (Junior blood group) Mus musculus 71-76 23732991-3 2013 Previously, we reported that the anticancer drug, sunitinib, an inhibitor of VEGF-R and PDGF-R, has off-target effects against both PKR and RNase L. Here we show that combining sunitinib treatments with infection by an oncolytic virus, vesicular stomatitis virus (VSV), led to the elimination of prostate, breast, and kidney malignant tumors in mice. Sunitinib 50-59 eukaryotic translation initiation factor 2-alpha kinase 2 Mus musculus 132-135 23732991-3 2013 Previously, we reported that the anticancer drug, sunitinib, an inhibitor of VEGF-R and PDGF-R, has off-target effects against both PKR and RNase L. Here we show that combining sunitinib treatments with infection by an oncolytic virus, vesicular stomatitis virus (VSV), led to the elimination of prostate, breast, and kidney malignant tumors in mice. Sunitinib 177-186 eukaryotic translation initiation factor 2-alpha kinase 2 Mus musculus 132-135 23732991-5 2013 In prostate tumors excised from treated mice, sunitinib decreased levels of the phosphorylated form of translation initiation factor, eIF2-alpha, a substrate of PKR, by 10-fold while increasing median viral titers by 23-fold. Sunitinib 46-55 eukaryotic translation initiation factor 2-alpha kinase 2 Mus musculus 134-144 23732991-5 2013 In prostate tumors excised from treated mice, sunitinib decreased levels of the phosphorylated form of translation initiation factor, eIF2-alpha, a substrate of PKR, by 10-fold while increasing median viral titers by 23-fold. Sunitinib 46-55 eukaryotic translation initiation factor 2-alpha kinase 2 Mus musculus 161-164 23578198-0 2013 Sunitinib induces cellular senescence via p53/Dec1 activation in renal cell carcinoma cells. Sunitinib 0-9 tumor protein p53 Homo sapiens 42-45 24003340-14 2013 Although not fully evaluated in pNETs, biomarkers associated with response to sunitinib in several tumor types include soluble vascular endothelial growth factor receptor 2 and 3, interleukin 8, and stromal cell-derived factor 1alpha. Sunitinib 78-87 C-X-C motif chemokine ligand 8 Homo sapiens 180-193 23964934-8 2013 Patients treated with sunitinib, as compared with those treated with pazopanib, had a higher incidence of fatigue (63% vs. 55%), the hand-foot syndrome (50% vs. 29%), and thrombocytopenia (78% vs. 41%); patients treated with pazopanib had a higher incidence of increased levels of alanine aminotransferase (60%, vs. 43% with sunitinib). Sunitinib 22-31 glutamic--pyruvic transaminase Homo sapiens 281-305 23887605-9 2013 RESULTS: We found that sunitinib treatment for 24 h triggers incomplete autophagy, impairs cathepsin B activation and stimulates a lysosomal-dependent necrosis. Sunitinib 23-32 cathepsin B Homo sapiens 91-102 23839492-3 2013 RESULTS: We demonstrated that, as single agents, sunitinib, sorafenib and everolimus share similar activity in inhibiting cell proliferation, signal transduction and vascular endothelial growth factor (VEGF) secretion in different RCC models, both in vitro and in tumour xenografts. Sunitinib 49-58 vascular endothelial growth factor A Homo sapiens 166-200 23839492-3 2013 RESULTS: We demonstrated that, as single agents, sunitinib, sorafenib and everolimus share similar activity in inhibiting cell proliferation, signal transduction and vascular endothelial growth factor (VEGF) secretion in different RCC models, both in vitro and in tumour xenografts. Sunitinib 49-58 vascular endothelial growth factor A Homo sapiens 202-206 23839492-5 2013 Inability by sunitinib to persistently inhibit HIF-1, VEGF and pMAPK anticipated treatment resistance in xenografted tumours. Sunitinib 13-22 hypoxia inducible factor 1 subunit alpha Homo sapiens 47-52 23839492-5 2013 Inability by sunitinib to persistently inhibit HIF-1, VEGF and pMAPK anticipated treatment resistance in xenografted tumours. Sunitinib 13-22 vascular endothelial growth factor A Homo sapiens 54-58 23839492-6 2013 After first-line sunitinib, second-line treatment with everolimus was more effective than either sorafenib or rechallenge with sunitinib in interfering with signalling proteins, VEGF and interleukin-8, translating into a significant advantage in tumour growth inhibition and mice survival. Sunitinib 127-136 vascular endothelial growth factor A Mus musculus 178-182 23839492-6 2013 After first-line sunitinib, second-line treatment with everolimus was more effective than either sorafenib or rechallenge with sunitinib in interfering with signalling proteins, VEGF and interleukin-8, translating into a significant advantage in tumour growth inhibition and mice survival. Sunitinib 127-136 chemokine (C-X-C motif) ligand 15 Mus musculus 187-200 23898066-1 2013 The present study investigated the effect of the H2 antagonist cimetidine on the pharmacokinetics of a multi-targeted receptor tyrosine kinase (RTK) inhibitor, sunitinib, in Sprague-Dawley (SD) rats and Eisai hyperbilirubinemic mutant rats (EHBR) lacking the efflux transporter, ATP-binding cassette C2 protein (ABCC2). Sunitinib 160-169 erb-b2 receptor tyrosine kinase 4 Rattus norvegicus 118-142 23898066-1 2013 The present study investigated the effect of the H2 antagonist cimetidine on the pharmacokinetics of a multi-targeted receptor tyrosine kinase (RTK) inhibitor, sunitinib, in Sprague-Dawley (SD) rats and Eisai hyperbilirubinemic mutant rats (EHBR) lacking the efflux transporter, ATP-binding cassette C2 protein (ABCC2). Sunitinib 160-169 erb-b2 receptor tyrosine kinase 4 Rattus norvegicus 144-147 23898066-7 2013 Furthermore, the contribution of ABCC2 to the biliary excretion of sunitinib was also examined in SD rats and EHBR. Sunitinib 67-76 ATP binding cassette subfamily C member 2 Rattus norvegicus 33-38 23898066-9 2013 These findings indicate that co-administration of cimetidine alters the pharmacokinetics of sunitinib probably due to inhibition of CYP3A4, suggesting the possibility that cimetidine should be used carefully for patients with cancer being treated with sunitinib therapy. Sunitinib 92-101 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 132-138 23578198-0 2013 Sunitinib induces cellular senescence via p53/Dec1 activation in renal cell carcinoma cells. Sunitinib 0-9 deleted in esophageal cancer 1 Homo sapiens 46-50 23578198-2 2013 Our present study revealed that sunitinib-treated RCC cells exhibit senescence characteristics including increased SA-beta-gal activity, DcR2 and Dec1 expression, and senescence-associated secretary phenotype (SASP) such as proinflammatory cytokines interleukin (IL)-1alpha, IL-6 and IL-8 secretion. Sunitinib 32-41 C-C motif chemokine receptor 6 Homo sapiens 137-141 23578198-2 2013 Our present study revealed that sunitinib-treated RCC cells exhibit senescence characteristics including increased SA-beta-gal activity, DcR2 and Dec1 expression, and senescence-associated secretary phenotype (SASP) such as proinflammatory cytokines interleukin (IL)-1alpha, IL-6 and IL-8 secretion. Sunitinib 32-41 deleted in esophageal cancer 1 Homo sapiens 146-150 23578198-2 2013 Our present study revealed that sunitinib-treated RCC cells exhibit senescence characteristics including increased SA-beta-gal activity, DcR2 and Dec1 expression, and senescence-associated secretary phenotype (SASP) such as proinflammatory cytokines interleukin (IL)-1alpha, IL-6 and IL-8 secretion. Sunitinib 32-41 interleukin 1 alpha Homo sapiens 250-273 23578198-2 2013 Our present study revealed that sunitinib-treated RCC cells exhibit senescence characteristics including increased SA-beta-gal activity, DcR2 and Dec1 expression, and senescence-associated secretary phenotype (SASP) such as proinflammatory cytokines interleukin (IL)-1alpha, IL-6 and IL-8 secretion. Sunitinib 32-41 interleukin 6 Homo sapiens 275-279 23578198-2 2013 Our present study revealed that sunitinib-treated RCC cells exhibit senescence characteristics including increased SA-beta-gal activity, DcR2 and Dec1 expression, and senescence-associated secretary phenotype (SASP) such as proinflammatory cytokines interleukin (IL)-1alpha, IL-6 and IL-8 secretion. Sunitinib 32-41 C-X-C motif chemokine ligand 8 Homo sapiens 284-288 23578198-4 2013 Mechanistic investigations indicated that therapy-induced senescence (TIS) following sunitinib treatment mainly attributed to p53/Dec1 signaling activation mediated by Raf-1/NF-kappaB inhibition in vitro. Sunitinib 85-94 tumor protein p53 Homo sapiens 126-129 23578198-4 2013 Mechanistic investigations indicated that therapy-induced senescence (TIS) following sunitinib treatment mainly attributed to p53/Dec1 signaling activation mediated by Raf-1/NF-kappaB inhibition in vitro. Sunitinib 85-94 deleted in esophageal cancer 1 Homo sapiens 130-134 23578198-4 2013 Mechanistic investigations indicated that therapy-induced senescence (TIS) following sunitinib treatment mainly attributed to p53/Dec1 signaling activation mediated by Raf-1/NF-kappaB inhibition in vitro. Sunitinib 85-94 Raf-1 proto-oncogene, serine/threonine kinase Homo sapiens 168-173 23578198-4 2013 Mechanistic investigations indicated that therapy-induced senescence (TIS) following sunitinib treatment mainly attributed to p53/Dec1 signaling activation mediated by Raf-1/NF-kappaB inhibition in vitro. Sunitinib 85-94 nuclear factor kappa B subunit 1 Homo sapiens 174-183 23578198-5 2013 Importantly, in vivo study showed tumor growth inhibition and prolonged overall survival were associated with increased p53 and Dec1 expression, decreased Raf-1 and Ki67 staining, and upregulated SA-beta-gal activity after sunitinib treatment. Sunitinib 223-232 Raf-1 proto-oncogene, serine/threonine kinase Homo sapiens 155-160 23578198-7 2013 Taken together, our findings suggested that sunitinib treatment performance could be attributable to TIS, depending on p53/Dec1 activation via inhibited Raf-1/nuclear factor (NF)-kappaB activity. Sunitinib 44-53 tumor protein p53 Homo sapiens 119-122 23578198-7 2013 Taken together, our findings suggested that sunitinib treatment performance could be attributable to TIS, depending on p53/Dec1 activation via inhibited Raf-1/nuclear factor (NF)-kappaB activity. Sunitinib 44-53 deleted in esophageal cancer 1 Homo sapiens 123-127 23578198-7 2013 Taken together, our findings suggested that sunitinib treatment performance could be attributable to TIS, depending on p53/Dec1 activation via inhibited Raf-1/nuclear factor (NF)-kappaB activity. Sunitinib 44-53 Raf-1 proto-oncogene, serine/threonine kinase Homo sapiens 153-158 23674235-6 2013 Approximately 50 % of patients with malignant PHs and SPGs have been found to carry hereditary germline mutations in the succinate dehydrogenase subunit B gene (SDHB), and anti-angiogenic agents such as sunitinib have been found to potentially play a role in the treatment of malignant disease, especially in patients with SDHB mutations. Sunitinib 203-212 succinate dehydrogenase complex iron sulfur subunit B Homo sapiens 161-165 23674235-6 2013 Approximately 50 % of patients with malignant PHs and SPGs have been found to carry hereditary germline mutations in the succinate dehydrogenase subunit B gene (SDHB), and anti-angiogenic agents such as sunitinib have been found to potentially play a role in the treatment of malignant disease, especially in patients with SDHB mutations. Sunitinib 203-212 succinate dehydrogenase complex iron sulfur subunit B Homo sapiens 323-327 23305097-8 2013 After sunitinib treatment, the expression levels of phosphorylated Akt and p44/42 mitogen-activated protein kinase in ACHN/P, but not those in ACHN/R, were significantly inhibited. Sunitinib 6-15 interferon induced protein 44 Homo sapiens 75-78 23880112-10 2013 The NF-kappaB inhibitors dicoumarol, SN50 and BAY11-7085 were employed to assess the role of NF-kappaB in sunitinib-mediated effects on neuronal survival as well as COX2 and NOS2 expression. Sunitinib 106-115 nuclear factor kappa B subunit 1 Homo sapiens 93-102 23880112-12 2013 Exposure of neurons to sunitinib also induced an increase in the expression of NF-kappaB, COX2 and NOS2. Sunitinib 23-32 nuclear factor kappa B subunit 1 Homo sapiens 79-88 23880112-12 2013 Exposure of neurons to sunitinib also induced an increase in the expression of NF-kappaB, COX2 and NOS2. Sunitinib 23-32 mitochondrially encoded cytochrome c oxidase II Homo sapiens 90-94 23880112-12 2013 Exposure of neurons to sunitinib also induced an increase in the expression of NF-kappaB, COX2 and NOS2. Sunitinib 23-32 nitric oxide synthase 2 Homo sapiens 99-103 23880112-13 2013 Inhibiting NF-kappaB blunted the increase in cell survival and decrease in cell death evoked by sunitinib. Sunitinib 96-105 nuclear factor kappa B subunit 1 Homo sapiens 11-20 23880112-14 2013 Treatment of cell cultures with both sunitinib and NF-kappaB inhibitors mitigated the increase in COX2 and NOS2 caused by sunitinib. Sunitinib 37-46 mitochondrially encoded cytochrome c oxidase II Homo sapiens 98-102 23880112-14 2013 Treatment of cell cultures with both sunitinib and NF-kappaB inhibitors mitigated the increase in COX2 and NOS2 caused by sunitinib. Sunitinib 37-46 nitric oxide synthase 2 Homo sapiens 107-111 23880112-14 2013 Treatment of cell cultures with both sunitinib and NF-kappaB inhibitors mitigated the increase in COX2 and NOS2 caused by sunitinib. Sunitinib 122-131 nuclear factor kappa B subunit 1 Homo sapiens 51-60 23880112-14 2013 Treatment of cell cultures with both sunitinib and NF-kappaB inhibitors mitigated the increase in COX2 and NOS2 caused by sunitinib. Sunitinib 122-131 mitochondrially encoded cytochrome c oxidase II Homo sapiens 98-102 23880112-14 2013 Treatment of cell cultures with both sunitinib and NF-kappaB inhibitors mitigated the increase in COX2 and NOS2 caused by sunitinib. Sunitinib 122-131 nitric oxide synthase 2 Homo sapiens 107-111 23880112-15 2013 CONCLUSIONS: Sunitinib increases neuronal survival and this neurotrophic effect is mediated by NF-kappaB. Sunitinib 13-22 nuclear factor kappa B subunit 1 Homo sapiens 95-104 23880112-16 2013 Also, the inflammatory proteins COX2 and NOS2 are upregulated by sunitinib in an NF-kappaB-dependent manner. Sunitinib 65-74 mitochondrially encoded cytochrome c oxidase II Homo sapiens 32-36 23880112-16 2013 Also, the inflammatory proteins COX2 and NOS2 are upregulated by sunitinib in an NF-kappaB-dependent manner. Sunitinib 65-74 nitric oxide synthase 2 Homo sapiens 41-45 23880112-16 2013 Also, the inflammatory proteins COX2 and NOS2 are upregulated by sunitinib in an NF-kappaB-dependent manner. Sunitinib 65-74 nuclear factor kappa B subunit 1 Homo sapiens 81-90 23842041-11 2013 CONCLUSION: In HCC, MRP may be a more sensitive biomarker in predicting early response and PFS following sunitinib than RECIST and mRECIST. Sunitinib 105-114 ATP binding cassette subfamily C member 1 Homo sapiens 20-23 23749551-0 2013 Effect of P-glycoprotein and breast cancer resistance protein inhibition on the pharmacokinetics of sunitinib in rats. Sunitinib 100-109 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 10-24 23749551-1 2013 The aim of this study was to elucidate the roles of P-glycoprotein (P-gp/ABCB1) and breast cancer resistance protein (BCRP/ABCG2) in the plasma concentration, biliary excretion, and distribution to the liver, kidney, and brain of sunitinib. Sunitinib 230-239 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 52-66 23749551-1 2013 The aim of this study was to elucidate the roles of P-glycoprotein (P-gp/ABCB1) and breast cancer resistance protein (BCRP/ABCG2) in the plasma concentration, biliary excretion, and distribution to the liver, kidney, and brain of sunitinib. Sunitinib 230-239 phosphoglycolate phosphatase Rattus norvegicus 68-72 23749551-1 2013 The aim of this study was to elucidate the roles of P-glycoprotein (P-gp/ABCB1) and breast cancer resistance protein (BCRP/ABCG2) in the plasma concentration, biliary excretion, and distribution to the liver, kidney, and brain of sunitinib. Sunitinib 230-239 ATP binding cassette subfamily B member 1A Rattus norvegicus 73-78 23749551-1 2013 The aim of this study was to elucidate the roles of P-glycoprotein (P-gp/ABCB1) and breast cancer resistance protein (BCRP/ABCG2) in the plasma concentration, biliary excretion, and distribution to the liver, kidney, and brain of sunitinib. Sunitinib 230-239 ATP binding cassette subfamily G member 2 Rattus norvegicus 123-128 23749551-7 2013 These results demonstrate that plasma concentrations of sunitinib and the biliary excretion and distribution to the kidney, liver, and brain of sunitinib are influenced by pharmacologic inhibition of P-gp and/or BCRP. Sunitinib 56-65 phosphoglycolate phosphatase Rattus norvegicus 200-204 23749551-7 2013 These results demonstrate that plasma concentrations of sunitinib and the biliary excretion and distribution to the kidney, liver, and brain of sunitinib are influenced by pharmacologic inhibition of P-gp and/or BCRP. Sunitinib 144-153 phosphoglycolate phosphatase Rattus norvegicus 200-204 23880112-0 2013 Sunitinib enhances neuronal survival in vitro via NF-kappaB-mediated signaling and expression of cyclooxygenase-2 and inducible nitric oxide synthase. Sunitinib 0-9 nuclear factor kappa B subunit 1 Homo sapiens 50-59 23880112-0 2013 Sunitinib enhances neuronal survival in vitro via NF-kappaB-mediated signaling and expression of cyclooxygenase-2 and inducible nitric oxide synthase. Sunitinib 0-9 prostaglandin-endoperoxide synthase 2 Homo sapiens 97-149 23880112-9 2013 The ability of sunitinib to affect NF-kappaB, COX2 and NOS2 expression was determined by western blot. Sunitinib 15-24 nuclear factor kappa B subunit 1 Homo sapiens 35-44 23880112-9 2013 The ability of sunitinib to affect NF-kappaB, COX2 and NOS2 expression was determined by western blot. Sunitinib 15-24 mitochondrially encoded cytochrome c oxidase II Homo sapiens 46-50 23880112-9 2013 The ability of sunitinib to affect NF-kappaB, COX2 and NOS2 expression was determined by western blot. Sunitinib 15-24 nitric oxide synthase 2 Homo sapiens 55-59 23305097-8 2013 After sunitinib treatment, the expression levels of phosphorylated Akt and p44/42 mitogen-activated protein kinase in ACHN/P, but not those in ACHN/R, were significantly inhibited. Sunitinib 6-15 La ribonucleoprotein 6, translational regulator Homo sapiens 118-124 23601669-2 2013 Sunitinib, sorafenib and pazopanib were the first generation of vascular endothelial growth factor receptor (VEGFR) tyrosine kinase inhibitors (TKIs) to have significant clinical activity in metastatic clear cell RCC. Sunitinib 0-9 kinase insert domain receptor Homo sapiens 64-107 23811706-1 2013 BACKGROUND: Therapy for metastatic kidney cancer is actively evolving, particularly in the results of registration drug trials that have led to the approval of vascular endothelial growth factor pathway drugs such as sorafenib, sunitinib, pazopanib, bevacizumab, and axitinib, with focus on patients with good- or intermediate-risk criteria and clear cell histology. Sunitinib 228-237 vascular endothelial growth factor A Homo sapiens 160-194 23867518-4 2013 Although high-dose interleukin 2 immunotherapy has been superseded as first-line therapy for RCC by promiscuous receptor tyrosine kinase inhibitors (rTKIs) such as sunitinib, sunitinib itself is a potent immunoadjunct in animal tumor models. Sunitinib 175-184 interleukin 2 Homo sapiens 19-32 23867518-8 2013 Although rTKIs like sunitinib have a remarkable capacity to deplete MDSCs and restore normal T-cell function in peripheral body compartments such as the bloodstream and the spleen, such rTKIs are effective only against MDSCs, which are engaged in phospho-STAT3-dependent programming (pSTAT3+). Sunitinib 20-29 signal transducer and activator of transcription 3 Homo sapiens 255-260 23601669-2 2013 Sunitinib, sorafenib and pazopanib were the first generation of vascular endothelial growth factor receptor (VEGFR) tyrosine kinase inhibitors (TKIs) to have significant clinical activity in metastatic clear cell RCC. Sunitinib 0-9 kinase insert domain receptor Homo sapiens 109-114 23582185-6 2013 Sublines with an additional T670I c-Kit mutation showed resistance to imatinib, nilotinib and dasatinib, but responded to sunitinib. Sunitinib 122-131 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 34-39 23840364-5 2013 Our results showed that SU more effectively inhibited mutant KIT with secondary exon 13 or 14 mutations than those with secondary exon 17 mutations, as clinically indicated. Sunitinib 24-26 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 61-64 23818948-10 2013 The apoptosis-inducing effects of sunitinib, but not sorafenib, were enhanced when IGFR signaling activity was inhibited by NVP-AEW541 or IGFR knockdown. Sunitinib 34-43 insulin like growth factor 1 receptor Homo sapiens 83-87 23818948-10 2013 The apoptosis-inducing effects of sunitinib, but not sorafenib, were enhanced when IGFR signaling activity was inhibited by NVP-AEW541 or IGFR knockdown. Sunitinib 34-43 insulin like growth factor 1 receptor Homo sapiens 138-142 23576559-5 2013 Inhibition of VEGF signaling by anti-VEGF antibody or sunitinib in mice from the age of 14 to 17 weeks was accompanied by more intratumoral lymphatics, more tumor cells inside lymphatics, and more lymph node metastases. Sunitinib 54-63 vascular endothelial growth factor A Mus musculus 14-18 23818948-11 2013 Chk2 kinase activation was found contributory to the synergistic anti-tumor effects between sunitinib and IGFR inhibition. Sunitinib 92-101 checkpoint kinase 2 Homo sapiens 0-4 23459719-3 2013 The results of clinical trials have further shown that the VEGF pathway inhibitor sunitinib and the mTOR inhibitor everolimus have efficacy in patients with advanced pancreatic NETs. Sunitinib 82-91 vascular endothelial growth factor A Homo sapiens 59-63 24396670-0 2013 Transformation of nonfunctioning pancreatic neuroendocrine carcinoma cells into insulin producing cells after treatment with sunitinib. Sunitinib 125-134 insulin Homo sapiens 80-87 24396670-6 2013 Immunohistochemical stain of the metastatic liver mass demonstrated that the initially nonfunctioning neuroendocrine carcinoma cells had changed into insulin-producing cells after sunitinib therapy. Sunitinib 180-189 insulin Homo sapiens 150-157 24396670-8 2013 A nonfunctioning pancreatic neuroendocrine carcinoma was transformed into an insulin-producing tumor after treatment with sunitinib, causing endogenous hyperinsulinemia and severe hypoglycemia. Sunitinib 122-131 insulin Homo sapiens 77-84 23455880-11 2013 Future studies will have to clarify whether KRAS can be used to guide sunitinib treatment or-in general-a treatment with a multityrosine kinase inhibitor in mCRC. Sunitinib 70-79 KRAS proto-oncogene, GTPase Homo sapiens 44-48 23705634-1 2013 INTRODUCTION: Some inhibitors of tyrosine kinase, as imatinib, erlotinib and sunitinib have antihyperglycemic effects but the mechanisms are not totally clear. Sunitinib 77-86 TXK tyrosine kinase Homo sapiens 33-48 23448320-11 2013 WHAT IS NEW AND CONCLUSION: P-glycoprotein inhibition by sunitinib has been demonstrated. Sunitinib 57-66 ATP binding cassette subfamily B member 1 Homo sapiens 28-42 23455880-0 2013 KRAS allel-specific activity of sunitinib in an isogenic disease model of colorectal cancer. Sunitinib 32-41 KRAS proto-oncogene, GTPase Homo sapiens 0-4 23455880-1 2013 PURPOSE: To investigate the impact of different KRAS mutations on treatment with the tyrosine kinase inhibitor sunitinib in SW48 colorectal cancer cell line variants. Sunitinib 111-120 KRAS proto-oncogene, GTPase Homo sapiens 48-52 23455880-3 2013 In addition, the respective cell lines were tested for the effect of sunitinib on ERK/ELK phosphorylation, cell cycle, and cytotoxicity. Sunitinib 69-78 EPH receptor B2 Homo sapiens 82-85 23455880-4 2013 RESULTS: Compared to KRAS wt cells, all KRAS mutant variants were associated with resistance to sunitinib treatment. Sunitinib 96-105 KRAS proto-oncogene, GTPase Homo sapiens 21-25 23455880-4 2013 RESULTS: Compared to KRAS wt cells, all KRAS mutant variants were associated with resistance to sunitinib treatment. Sunitinib 96-105 KRAS proto-oncogene, GTPase Homo sapiens 40-44 23455880-6 2013 The reduction in ERK phosphorylation due to treatment with sunitinib was highest in G12V (89 %) mutant cells and lowest in G12A (24 %) mutant cells. Sunitinib 59-68 EPH receptor B2 Homo sapiens 17-20 23392356-0 2013 The secondary FLT3-ITD F691L mutation induces resistance to AC220 in FLT3-ITD+ AML but retains in vitro sensitivity to PKC412 and Sunitinib. Sunitinib 130-139 fms related receptor tyrosine kinase 3 Homo sapiens 14-18 23455880-7 2013 ELK phosphorylation was less decreased in all KRAS mutant variants compared to KRAS wt cells following sunitinib treatment. Sunitinib 103-112 KRAS proto-oncogene, GTPase Homo sapiens 46-50 23455880-7 2013 ELK phosphorylation was less decreased in all KRAS mutant variants compared to KRAS wt cells following sunitinib treatment. Sunitinib 103-112 KRAS proto-oncogene, GTPase Homo sapiens 79-83 23455880-9 2013 CONCLUSION: Our isogenic cell culture model suggests that KRAS mutations in SW48 colorectal cancer cells are linked to resistance to the multityrosine kinase inhibitor sunitinib. Sunitinib 168-177 KRAS proto-oncogene, GTPase Homo sapiens 58-62 23730410-11 2013 CONCLUSION: Ang-2 could potentially identify a patient population that might have a better PFS when under anti-angiogenic treatment, like the tyrosine kinase inhibitor sunitinib. Sunitinib 168-177 angiopoietin 2 Homo sapiens 12-17 23456621-0 2013 Secondary mutations of c-KIT contribute to acquired resistance to imatinib and decrease efficacy of sunitinib in Chinese patients with gastrointestinal stromal tumors. Sunitinib 100-109 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 23-28 23456621-1 2013 The aim of this study was to investigate the associations between secondary mutations of c-KIT/PDGFRalpha and acquired imatinib resistance or efficacy of sunitinib in Chinese patients with gastrointestinal stromal tumors (GISTs). Sunitinib 154-163 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 89-94 23456621-1 2013 The aim of this study was to investigate the associations between secondary mutations of c-KIT/PDGFRalpha and acquired imatinib resistance or efficacy of sunitinib in Chinese patients with gastrointestinal stromal tumors (GISTs). Sunitinib 154-163 platelet derived growth factor receptor alpha Homo sapiens 95-105 23456621-8 2013 Secondary mutations of c-KIT were significantly associated with acquired resistance to imatinib in Chinese GIST patients, and whether secondary mutations of c-KIT could influence the efficacy of sunitinib needed to be further investigated. Sunitinib 195-204 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 157-162 23539200-6 2013 A significant decrease in the Th1/Th2 ratio was seen after sunitinib treatment only in the PFS-short group, suggesting a shift toward Th2 that down-regulates host immunity. Sunitinib 59-68 negative elongation factor complex member C/D Homo sapiens 30-33 23720580-6 2013 Sunitinib-induced pericyte depletion and coronary microvascular dysfunction are recapitulated by CP-673451, a structurally distinct PDGFR inhibitor, confirming the role of PDGFR in pericyte survival. Sunitinib 0-9 platelet derived growth factor receptor beta Homo sapiens 132-137 23720580-6 2013 Sunitinib-induced pericyte depletion and coronary microvascular dysfunction are recapitulated by CP-673451, a structurally distinct PDGFR inhibitor, confirming the role of PDGFR in pericyte survival. Sunitinib 0-9 platelet derived growth factor receptor beta Homo sapiens 172-177 23522954-11 2013 Finally, we checked whether compensatory activation of VEGF signaling occurred after sunitinib addition. Sunitinib 85-94 vascular endothelial growth factor A Homo sapiens 55-59 23492365-8 2013 Notably, JMJD1A inhibition enhanced the antitumor effects of the anti-VEGF compound bevacizumab and the VEGFR/KDR inhibitor sunitinib. Sunitinib 124-133 lysine demethylase 3A Homo sapiens 9-15 23492365-8 2013 Notably, JMJD1A inhibition enhanced the antitumor effects of the anti-VEGF compound bevacizumab and the VEGFR/KDR inhibitor sunitinib. Sunitinib 124-133 kinase insert domain receptor Homo sapiens 104-109 23492365-8 2013 Notably, JMJD1A inhibition enhanced the antitumor effects of the anti-VEGF compound bevacizumab and the VEGFR/KDR inhibitor sunitinib. Sunitinib 124-133 kinase insert domain receptor Homo sapiens 110-113 23288144-0 2013 Sunitinib, a tyrosine kinase inhibitor, induces cytochrome P450 1A1 gene in human breast cancer MCF7 cells through ligand-independent aryl hydrocarbon receptor activation. Sunitinib 0-9 cytochrome P450 family 1 subfamily A member 1 Homo sapiens 48-67 23288144-0 2013 Sunitinib, a tyrosine kinase inhibitor, induces cytochrome P450 1A1 gene in human breast cancer MCF7 cells through ligand-independent aryl hydrocarbon receptor activation. Sunitinib 0-9 aryl hydrocarbon receptor Homo sapiens 134-159 23580490-8 2013 Reduced F4/80+ cell infiltration was evidenced in sunitinib-treated mice and was associated to decreased VEGF-A (by 50%, P < 0.01) and VEGF-C (by 35%, P < 0.01) expressions, while VEGF-D and sVEGFR-2 expressions were not affected. Sunitinib 50-59 vascular endothelial growth factor A Mus musculus 105-111 23580490-8 2013 Reduced F4/80+ cell infiltration was evidenced in sunitinib-treated mice and was associated to decreased VEGF-A (by 50%, P < 0.01) and VEGF-C (by 35%, P < 0.01) expressions, while VEGF-D and sVEGFR-2 expressions were not affected. Sunitinib 50-59 vascular endothelial growth factor C Mus musculus 138-144 23580490-8 2013 Reduced F4/80+ cell infiltration was evidenced in sunitinib-treated mice and was associated to decreased VEGF-A (by 50%, P < 0.01) and VEGF-C (by 35%, P < 0.01) expressions, while VEGF-D and sVEGFR-2 expressions were not affected. Sunitinib 50-59 vascular endothelial growth factor D Mus musculus 186-192 23580490-8 2013 Reduced F4/80+ cell infiltration was evidenced in sunitinib-treated mice and was associated to decreased VEGF-A (by 50%, P < 0.01) and VEGF-C (by 35%, P < 0.01) expressions, while VEGF-D and sVEGFR-2 expressions were not affected. Sunitinib 50-59 kinase insert domain protein receptor Mus musculus 197-205 23580490-11 2013 Mechanistically, sunitinib blocked VEGFR-2 phosphorylation induced by VEGF-A released by macrophages. Sunitinib 17-26 kinase insert domain protein receptor Mus musculus 35-42 23580490-11 2013 Mechanistically, sunitinib blocked VEGFR-2 phosphorylation induced by VEGF-A released by macrophages. Sunitinib 17-26 vascular endothelial growth factor A Mus musculus 70-76 23206420-11 2013 CONCLUSIONS: In our study microRNA-141 down-regulation driven epithelial-to-mesenchymal transition in clear cell renal cell carcinoma was linked to an unfavorable response to sunitinib therapy. Sunitinib 175-184 microRNA 141 Homo sapiens 26-38 23206420-13 2013 Future experiments should be done in vivo to see whether microRNA-141 driven reversal of epithelial-to-mesenchymal transition could affect the efficacy of sunitinib treatment. Sunitinib 155-164 microRNA 141 Homo sapiens 57-69 23206420-0 2013 A possible role for microRNA-141 down-regulation in sunitinib resistant metastatic clear cell renal cell carcinoma through induction of epithelial-to-mesenchymal transition and hypoxia resistance. Sunitinib 52-61 microRNA 141 Homo sapiens 20-32 21478036-0 2013 Expression level of vascular endothelial growth factor receptor-2 in radical nephrectomy specimens as a prognostic predictor in patients with metastatic renal cell carcinoma treated with sunitinib. Sunitinib 187-196 kinase insert domain receptor Homo sapiens 20-65 23206420-8 2013 RESULTS: Compared to good responders, microRNA-141 was significantly down-regulated in tumors of poor responders to sunitinib. Sunitinib 116-125 microRNA 141 Homo sapiens 38-50 23819366-6 2013 Sunitinib was administered following Interferon alpha (IFN-alpha) therapy; however, the lung metastases became larger, so administration of everolimus at 10 mg/day was commenced. Sunitinib 0-9 interferon alpha 1 Homo sapiens 55-64 21478036-4 2013 RESULTS: Of several factors examined, tumor grade and the expression level of VEGFR-2 were shown to have significant impacts on response to sunitinib in these 40 patients. Sunitinib 140-149 kinase insert domain receptor Homo sapiens 78-85 21478036-8 2013 CONCLUSIONS: Collectively, these findings suggest that it would be useful to consider expression levels of potential molecular markers, particularly VEGFR-2, as well as conventional clinical parameters to select metastatic RCC patients likely to benefit from treatment with sunitinib. Sunitinib 274-283 kinase insert domain receptor Homo sapiens 149-156 23180436-5 2013 Sunitinib induced G1 phase arrest associated with decreased cyclin D1, cyclin D3, and cyclin-dependent kinase (Cdk)2 and increased p27(Kip1), pRb1, and p130/Rb2 expression and phosphorylated activation of protein kinase C alpha and beta (PKCalpha/beta). Sunitinib 0-9 cyclin dependent kinase inhibitor 1B Homo sapiens 135-139 23454556-4 2013 We performed in vitro experiments to study whether Sunitinib, a platelet-derived growth factor (PDGF) and vascular endothelial growth factor (VEGF) RTKs" inhibitor, could block both activated HSC functions and angiogenesis and thus prevent the progression of cirrhotic liver to hepatocellular carcinoma. Sunitinib 51-60 vascular endothelial growth factor A Homo sapiens 106-140 23454556-4 2013 We performed in vitro experiments to study whether Sunitinib, a platelet-derived growth factor (PDGF) and vascular endothelial growth factor (VEGF) RTKs" inhibitor, could block both activated HSC functions and angiogenesis and thus prevent the progression of cirrhotic liver to hepatocellular carcinoma. Sunitinib 51-60 vascular endothelial growth factor A Homo sapiens 142-146 23454556-8 2013 In conclusion, the present study demonstrates that the RTK inhibitor Sunitinib blocks the activation of HSC and angiogenesis suggesting its potential as a drug candidate in pathological conditions like liver fibrosis and hepatocellular carcinoma. Sunitinib 69-78 ret proto-oncogene Homo sapiens 55-58 23544171-12 2013 Importantly, phospho-RTK arrays revealed novel targets for cediranib and sunitinib therapy. Sunitinib 73-82 ret proto-oncogene Homo sapiens 21-24 23511629-0 2013 VEGF and VEGFR polymorphisms affect clinical outcome in advanced renal cell carcinoma patients receiving first-line sunitinib. Sunitinib 116-125 vascular endothelial growth factor A Homo sapiens 0-4 23511629-0 2013 VEGF and VEGFR polymorphisms affect clinical outcome in advanced renal cell carcinoma patients receiving first-line sunitinib. Sunitinib 116-125 kinase insert domain receptor Homo sapiens 9-14 23378344-8 2013 Having identified PDGFR-beta to be regulated by FOXC2, we show that the U.S. Food and Drug Administration-approved PDGFR inhibitor, sunitinib, targets FOXC2-expressing tumor cells leading to reduced CSC and metastatic properties. Sunitinib 132-141 platelet derived growth factor receptor beta Homo sapiens 18-28 23378344-8 2013 Having identified PDGFR-beta to be regulated by FOXC2, we show that the U.S. Food and Drug Administration-approved PDGFR inhibitor, sunitinib, targets FOXC2-expressing tumor cells leading to reduced CSC and metastatic properties. Sunitinib 132-141 forkhead box C2 Homo sapiens 48-53 23378344-8 2013 Having identified PDGFR-beta to be regulated by FOXC2, we show that the U.S. Food and Drug Administration-approved PDGFR inhibitor, sunitinib, targets FOXC2-expressing tumor cells leading to reduced CSC and metastatic properties. Sunitinib 132-141 platelet derived growth factor receptor beta Homo sapiens 18-23 23378344-8 2013 Having identified PDGFR-beta to be regulated by FOXC2, we show that the U.S. Food and Drug Administration-approved PDGFR inhibitor, sunitinib, targets FOXC2-expressing tumor cells leading to reduced CSC and metastatic properties. Sunitinib 132-141 forkhead box C2 Homo sapiens 151-156 23462807-7 2013 CONCLUSION: Our results confirm former communications regarding the association between SNPs in ABCB1, NR1/2, NR1/3 and VEGFR3 and sunitinib outcome in clear-cell RCC. Sunitinib 131-140 ATP binding cassette subfamily B member 1 Homo sapiens 96-101 23462807-7 2013 CONCLUSION: Our results confirm former communications regarding the association between SNPs in ABCB1, NR1/2, NR1/3 and VEGFR3 and sunitinib outcome in clear-cell RCC. Sunitinib 131-140 glutamate ionotropic receptor NMDA type subunit 1 Homo sapiens 103-108 23462807-7 2013 CONCLUSION: Our results confirm former communications regarding the association between SNPs in ABCB1, NR1/2, NR1/3 and VEGFR3 and sunitinib outcome in clear-cell RCC. Sunitinib 131-140 glutamate ionotropic receptor NMDA type subunit 1 Homo sapiens 110-115 23462807-7 2013 CONCLUSION: Our results confirm former communications regarding the association between SNPs in ABCB1, NR1/2, NR1/3 and VEGFR3 and sunitinib outcome in clear-cell RCC. Sunitinib 131-140 fms related receptor tyrosine kinase 4 Homo sapiens 120-126 23497256-4 2013 RESULTS: Sunitinib treatment increased p53 levels in RCC xenografts and transiently induced the expression of p21(waf1), Noxa, and HDM2, the levels of which subsequently declined to baseline (or undetectable) with the emergence of sunitinib resistance. Sunitinib 9-18 tumor protein p53 Homo sapiens 39-42 23497256-4 2013 RESULTS: Sunitinib treatment increased p53 levels in RCC xenografts and transiently induced the expression of p21(waf1), Noxa, and HDM2, the levels of which subsequently declined to baseline (or undetectable) with the emergence of sunitinib resistance. Sunitinib 9-18 cyclin dependent kinase inhibitor 1A Homo sapiens 110-113 23497256-4 2013 RESULTS: Sunitinib treatment increased p53 levels in RCC xenografts and transiently induced the expression of p21(waf1), Noxa, and HDM2, the levels of which subsequently declined to baseline (or undetectable) with the emergence of sunitinib resistance. Sunitinib 9-18 cyclin dependent kinase inhibitor 1A Homo sapiens 114-118 23497256-4 2013 RESULTS: Sunitinib treatment increased p53 levels in RCC xenografts and transiently induced the expression of p21(waf1), Noxa, and HDM2, the levels of which subsequently declined to baseline (or undetectable) with the emergence of sunitinib resistance. Sunitinib 9-18 phorbol-12-myristate-13-acetate-induced protein 1 Homo sapiens 121-125 23497256-4 2013 RESULTS: Sunitinib treatment increased p53 levels in RCC xenografts and transiently induced the expression of p21(waf1), Noxa, and HDM2, the levels of which subsequently declined to baseline (or undetectable) with the emergence of sunitinib resistance. Sunitinib 9-18 MDM2 proto-oncogene Homo sapiens 131-135 23497256-7 2013 It also suppressed the expression of the chemokine SDF-1 (CXCL12) and the influx of CD11b+/Gr-1+ myeloid-derived suppressor cells (MDSC) otherwise induced by sunitinib. Sunitinib 158-167 integrin subunit alpha M Homo sapiens 84-89 23180436-5 2013 Sunitinib induced G1 phase arrest associated with decreased cyclin D1, cyclin D3, and cyclin-dependent kinase (Cdk)2 and increased p27(Kip1), pRb1, and p130/Rb2 expression and phosphorylated activation of protein kinase C alpha and beta (PKCalpha/beta). Sunitinib 0-9 RB transcriptional corepressor like 2 Homo sapiens 152-156 23180436-5 2013 Sunitinib induced G1 phase arrest associated with decreased cyclin D1, cyclin D3, and cyclin-dependent kinase (Cdk)2 and increased p27(Kip1), pRb1, and p130/Rb2 expression and phosphorylated activation of protein kinase C alpha and beta (PKCalpha/beta). Sunitinib 0-9 cyclin D1 Homo sapiens 60-69 23180436-5 2013 Sunitinib induced G1 phase arrest associated with decreased cyclin D1, cyclin D3, and cyclin-dependent kinase (Cdk)2 and increased p27(Kip1), pRb1, and p130/Rb2 expression and phosphorylated activation of protein kinase C alpha and beta (PKCalpha/beta). Sunitinib 0-9 cyclin D3 Homo sapiens 71-80 23180436-5 2013 Sunitinib induced G1 phase arrest associated with decreased cyclin D1, cyclin D3, and cyclin-dependent kinase (Cdk)2 and increased p27(Kip1), pRb1, and p130/Rb2 expression and phosphorylated activation of protein kinase C alpha and beta (PKCalpha/beta). Sunitinib 0-9 cyclin dependent kinase 2 Homo sapiens 111-116 23180436-5 2013 Sunitinib induced G1 phase arrest associated with decreased cyclin D1, cyclin D3, and cyclin-dependent kinase (Cdk)2 and increased p27(Kip1), pRb1, and p130/Rb2 expression and phosphorylated activation of protein kinase C alpha and beta (PKCalpha/beta). Sunitinib 0-9 interferon alpha inducible protein 27 Homo sapiens 131-134 23375402-3 2013 Here, we present a case report of TC with c-Kit mutation, who has relapsed after exposure to multiple lines of combination chemotherapy, but he has shown an impressive and long lasting response to sunitinib after imatinib failure. Sunitinib 197-206 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 42-47 23414694-15 2013 Responders to primary sunitinib who underwent CRN had better DSS and OS than patients who underwent primary CRN, followed by sunitinib. Sunitinib 22-31 crooked neck pre-mRNA splicing factor 1 Homo sapiens 46-49 23180436-5 2013 Sunitinib induced G1 phase arrest associated with decreased cyclin D1, cyclin D3, and cyclin-dependent kinase (Cdk)2 and increased p27(Kip1), pRb1, and p130/Rb2 expression and phosphorylated activation of protein kinase C alpha and beta (PKCalpha/beta). Sunitinib 0-9 proline rich protein BstNI subfamily 1 Homo sapiens 142-146 23180436-5 2013 Sunitinib induced G1 phase arrest associated with decreased cyclin D1, cyclin D3, and cyclin-dependent kinase (Cdk)2 and increased p27(Kip1), pRb1, and p130/Rb2 expression and phosphorylated activation of protein kinase C alpha and beta (PKCalpha/beta). Sunitinib 0-9 RB transcriptional corepressor like 2 Homo sapiens 157-160 23180436-5 2013 Sunitinib induced G1 phase arrest associated with decreased cyclin D1, cyclin D3, and cyclin-dependent kinase (Cdk)2 and increased p27(Kip1), pRb1, and p130/Rb2 expression and phosphorylated activation of protein kinase C alpha and beta (PKCalpha/beta). Sunitinib 0-9 protein kinase C alpha Homo sapiens 205-227 23180436-5 2013 Sunitinib induced G1 phase arrest associated with decreased cyclin D1, cyclin D3, and cyclin-dependent kinase (Cdk)2 and increased p27(Kip1), pRb1, and p130/Rb2 expression and phosphorylated activation of protein kinase C alpha and beta (PKCalpha/beta). Sunitinib 0-9 protein kinase C alpha Homo sapiens 238-251 23180436-6 2013 Selective PKCalpha/beta inhibitor treatment abolished sunitinib-elicited AML differentiation, suggesting that PKCalpha/beta may underlie sunitinib-induced monocytic differentiation. Sunitinib 54-63 protein kinase C alpha Homo sapiens 10-23 23180436-6 2013 Selective PKCalpha/beta inhibitor treatment abolished sunitinib-elicited AML differentiation, suggesting that PKCalpha/beta may underlie sunitinib-induced monocytic differentiation. Sunitinib 54-63 protein kinase C alpha Homo sapiens 110-123 23180436-6 2013 Selective PKCalpha/beta inhibitor treatment abolished sunitinib-elicited AML differentiation, suggesting that PKCalpha/beta may underlie sunitinib-induced monocytic differentiation. Sunitinib 137-146 protein kinase C alpha Homo sapiens 10-23 23180436-6 2013 Selective PKCalpha/beta inhibitor treatment abolished sunitinib-elicited AML differentiation, suggesting that PKCalpha/beta may underlie sunitinib-induced monocytic differentiation. Sunitinib 137-146 protein kinase C alpha Homo sapiens 110-123 30349606-6 2013 Motesanib, sorafenib, vandetanib, sunitinib, lenvatinib, imatinib and cabozantinib are multi-kinase inhibitors that have the ability of inhibiting the rearranged during transection (RET) and vascular endothelial growth factor receptor (VEGFR), and other kinases, and have been used in advanced differentiated thyroid carcinoma. Sunitinib 34-43 ret proto-oncogene Homo sapiens 182-185 23507600-8 2013 Sunitinib is a tyrosine kinase inhibitor for multi-targets including VEGFR, PDGFR, c-kit et. Sunitinib 0-9 kinase insert domain receptor Homo sapiens 69-74 23507600-8 2013 Sunitinib is a tyrosine kinase inhibitor for multi-targets including VEGFR, PDGFR, c-kit et. Sunitinib 0-9 platelet derived growth factor receptor beta Homo sapiens 76-81 23507600-8 2013 Sunitinib is a tyrosine kinase inhibitor for multi-targets including VEGFR, PDGFR, c-kit et. Sunitinib 0-9 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 83-88 23114714-8 2013 Overall, our results suggest a role for Cav-1 as a biomarker of response to both dasatinib and sunitinib treatment and as a therapeutic target in prostate cancer. Sunitinib 95-104 caveolin 1, caveolae protein Mus musculus 40-45 23201752-0 2013 MicroRNA-21 silencing enhances the cytotoxic effect of the antiangiogenic drug sunitinib in glioblastoma. Sunitinib 79-88 microRNA 21 Homo sapiens 0-11 23201752-6 2013 Finally, we demonstrate for the first time that miR-21 silencing enhances the antitumoral effect of the tyrosine kinase inhibitor sunitinib, whereas no therapeutic benefit is observed when coupling miR-21 silencing with the first-line drug temozolomide. Sunitinib 130-139 microRNA 21 Homo sapiens 48-54 23536339-12 2013 The sub-group analysis of two protocols of sunitinib administration showed that the incidence of diarrhea and hand-foot syndrome were higher in 50 mg/d (4/2) group than those in 37.5 mg/d CDD group (P=0.027, P=0.048). Sunitinib 43-52 natriuretic peptide A Homo sapiens 188-191 23536339-16 2013 The protocol of sunitinib 37.5 mg/d CDD possesses better safety. Sunitinib 16-25 natriuretic peptide A Homo sapiens 36-39 23626551-0 2013 Targeting VEGF-VEGFR Pathway by Sunitinib in Peripheral Primitive Neuroectodermal Tumor, Paraganglioma and Epithelioid Hemangioendothelioma: Three Case Reports. Sunitinib 32-41 vascular endothelial growth factor A Homo sapiens 10-14 23626551-0 2013 Targeting VEGF-VEGFR Pathway by Sunitinib in Peripheral Primitive Neuroectodermal Tumor, Paraganglioma and Epithelioid Hemangioendothelioma: Three Case Reports. Sunitinib 32-41 kinase insert domain receptor Homo sapiens 15-20 23179081-5 2013 Although the precise mechanism of these interactions is not well understood, cytochrome P450 mono-oxygenase 3A4 (CYP3A4) is a key enzyme to inactivate both glucocorticoids and sunitinib. Sunitinib 176-185 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 77-111 23179081-5 2013 Although the precise mechanism of these interactions is not well understood, cytochrome P450 mono-oxygenase 3A4 (CYP3A4) is a key enzyme to inactivate both glucocorticoids and sunitinib. Sunitinib 176-185 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 113-119 23114714-2 2013 Treatment of both cell lines with either dasatinib or sunitinib reduced phosphorylation of PDGFR, VEGFR2, Akt, FAK, Src (dasatinib only) and Cav-1, and reduced cellular and secreted levels of Cav-1. Sunitinib 54-63 platelet derived growth factor receptor, beta polypeptide Mus musculus 91-96 23114714-2 2013 Treatment of both cell lines with either dasatinib or sunitinib reduced phosphorylation of PDGFR, VEGFR2, Akt, FAK, Src (dasatinib only) and Cav-1, and reduced cellular and secreted levels of Cav-1. Sunitinib 54-63 kinase insert domain protein receptor Mus musculus 98-104 23114714-2 2013 Treatment of both cell lines with either dasatinib or sunitinib reduced phosphorylation of PDGFR, VEGFR2, Akt, FAK, Src (dasatinib only) and Cav-1, and reduced cellular and secreted levels of Cav-1. Sunitinib 54-63 thymoma viral proto-oncogene 1 Mus musculus 106-109 23269235-2 2013 Previously, we demonstrated that sunitinib directly inhibited mTORC1 signaling in rat pheochromocytoma PC12 cells. Sunitinib 33-42 CREB regulated transcription coactivator 1 Mus musculus 62-68 23114714-2 2013 Treatment of both cell lines with either dasatinib or sunitinib reduced phosphorylation of PDGFR, VEGFR2, Akt, FAK, Src (dasatinib only) and Cav-1, and reduced cellular and secreted levels of Cav-1. Sunitinib 54-63 PTK2 protein tyrosine kinase 2 Mus musculus 111-114 23114714-2 2013 Treatment of both cell lines with either dasatinib or sunitinib reduced phosphorylation of PDGFR, VEGFR2, Akt, FAK, Src (dasatinib only) and Cav-1, and reduced cellular and secreted levels of Cav-1. Sunitinib 54-63 Rous sarcoma oncogene Mus musculus 116-119 23114714-2 2013 Treatment of both cell lines with either dasatinib or sunitinib reduced phosphorylation of PDGFR, VEGFR2, Akt, FAK, Src (dasatinib only) and Cav-1, and reduced cellular and secreted levels of Cav-1. Sunitinib 54-63 caveolin 1, caveolae protein Mus musculus 141-146 23114714-2 2013 Treatment of both cell lines with either dasatinib or sunitinib reduced phosphorylation of PDGFR, VEGFR2, Akt, FAK, Src (dasatinib only) and Cav-1, and reduced cellular and secreted levels of Cav-1. Sunitinib 54-63 caveolin 1, caveolae protein Mus musculus 192-197 23114714-6 2013 Serum Cav-1 levels were lower in dasatinib- and sunitinib-treated mice than they were in vehicle-treated mice, and correlated positively with tumor growth in dasatinib- and sunitinib-treated groups (r = 0.48, p = 0.031; r = 0.554, p = 0.0065, respectively), compared with vehicle controls. Sunitinib 48-57 caveolin 1, caveolae protein Mus musculus 6-11 23114714-6 2013 Serum Cav-1 levels were lower in dasatinib- and sunitinib-treated mice than they were in vehicle-treated mice, and correlated positively with tumor growth in dasatinib- and sunitinib-treated groups (r = 0.48, p = 0.031; r = 0.554, p = 0.0065, respectively), compared with vehicle controls. Sunitinib 173-182 caveolin 1, caveolae protein Mus musculus 6-11 23114714-7 2013 Cav-1 knockdown, in combination with dasatinib or sunitinib treatment in PC-3 cells, caused a greater reduction in the phosphorylation of PDGFR-beta and VEGFR2, and expression and secretion of PDGF-B and VEGF-A than that in PC-3 cells treated with dasatinib or sunitinib alone in control siRNA cells, suggesting that Cav-1 is involved in an autocrine pathway that is affected by these drugs. Sunitinib 50-59 caveolin 1, caveolae protein Mus musculus 0-5 23114714-7 2013 Cav-1 knockdown, in combination with dasatinib or sunitinib treatment in PC-3 cells, caused a greater reduction in the phosphorylation of PDGFR-beta and VEGFR2, and expression and secretion of PDGF-B and VEGF-A than that in PC-3 cells treated with dasatinib or sunitinib alone in control siRNA cells, suggesting that Cav-1 is involved in an autocrine pathway that is affected by these drugs. Sunitinib 50-59 platelet derived growth factor receptor, beta polypeptide Mus musculus 138-148 23114714-7 2013 Cav-1 knockdown, in combination with dasatinib or sunitinib treatment in PC-3 cells, caused a greater reduction in the phosphorylation of PDGFR-beta and VEGFR2, and expression and secretion of PDGF-B and VEGF-A than that in PC-3 cells treated with dasatinib or sunitinib alone in control siRNA cells, suggesting that Cav-1 is involved in an autocrine pathway that is affected by these drugs. Sunitinib 50-59 kinase insert domain protein receptor Mus musculus 153-159 23114714-7 2013 Cav-1 knockdown, in combination with dasatinib or sunitinib treatment in PC-3 cells, caused a greater reduction in the phosphorylation of PDGFR-beta and VEGFR2, and expression and secretion of PDGF-B and VEGF-A than that in PC-3 cells treated with dasatinib or sunitinib alone in control siRNA cells, suggesting that Cav-1 is involved in an autocrine pathway that is affected by these drugs. Sunitinib 50-59 platelet derived growth factor, B polypeptide Mus musculus 193-199 23114714-7 2013 Cav-1 knockdown, in combination with dasatinib or sunitinib treatment in PC-3 cells, caused a greater reduction in the phosphorylation of PDGFR-beta and VEGFR2, and expression and secretion of PDGF-B and VEGF-A than that in PC-3 cells treated with dasatinib or sunitinib alone in control siRNA cells, suggesting that Cav-1 is involved in an autocrine pathway that is affected by these drugs. Sunitinib 50-59 vascular endothelial growth factor A Mus musculus 204-210 23114714-7 2013 Cav-1 knockdown, in combination with dasatinib or sunitinib treatment in PC-3 cells, caused a greater reduction in the phosphorylation of PDGFR-beta and VEGFR2, and expression and secretion of PDGF-B and VEGF-A than that in PC-3 cells treated with dasatinib or sunitinib alone in control siRNA cells, suggesting that Cav-1 is involved in an autocrine pathway that is affected by these drugs. Sunitinib 50-59 caveolin 1, caveolae protein Mus musculus 317-322 23114714-7 2013 Cav-1 knockdown, in combination with dasatinib or sunitinib treatment in PC-3 cells, caused a greater reduction in the phosphorylation of PDGFR-beta and VEGFR2, and expression and secretion of PDGF-B and VEGF-A than that in PC-3 cells treated with dasatinib or sunitinib alone in control siRNA cells, suggesting that Cav-1 is involved in an autocrine pathway that is affected by these drugs. Sunitinib 261-270 caveolin 1, caveolae protein Mus musculus 0-5 23398161-1 2013 BACKGROUND: Thyroid dysfunction is a well-known adverse effect of sunitinib, a drug that targets multiple receptor tyrosine kinases, including vascular endothelial growth factor receptor (VEGFR). Sunitinib 66-75 kinase insert domain receptor Homo sapiens 143-186 23398161-1 2013 BACKGROUND: Thyroid dysfunction is a well-known adverse effect of sunitinib, a drug that targets multiple receptor tyrosine kinases, including vascular endothelial growth factor receptor (VEGFR). Sunitinib 66-75 kinase insert domain receptor Homo sapiens 188-193 23399884-7 2013 Some antiangiogenic drugs inhibiting the VEGFR2 receptor are successfully used in clinics for the treatment of several types of tumours in synergy with chemotherapy (e.g., Nexavar( ) from Bayer, Sutent( ) from Pfizer and Votrient( ) from GlaxoSmithKline, approved by the FDA in 2005, 2006 and 2009, respectively). Sunitinib 195-201 kinase insert domain receptor Homo sapiens 41-47 23127174-6 2013 The action of KIT kinase inhibitors, especially imatinib, sunitinib, dasatinib and PKC412, on different primary and secondary mutants is discussed. Sunitinib 58-67 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 14-17 23613413-2 2013 METHODS: Three different doses of sutent (D1= 1 muM, D2= 5 muM, D3= 10 muM) were administered to cells for 72 h to determine the optimal dose. Sunitinib 34-40 latexin Homo sapiens 48-51 23613413-2 2013 METHODS: Three different doses of sutent (D1= 1 muM, D2= 5 muM, D3= 10 muM) were administered to cells for 72 h to determine the optimal dose. Sunitinib 34-40 latexin Homo sapiens 59-62 23613413-2 2013 METHODS: Three different doses of sutent (D1= 1 muM, D2= 5 muM, D3= 10 muM) were administered to cells for 72 h to determine the optimal dose. Sunitinib 34-40 latexin Homo sapiens 59-62 22995920-2 2013 Here, we describe a case of TFE3-positive RCC in which metastatic relapse to the mediastinal lymph nodes and pulmonary nodules was treated with single-agent sunitinib, a multitargeted tyrosine inhibitor. Sunitinib 157-166 transcription factor binding to IGHM enhancer 3 Homo sapiens 28-32 23322198-0 2013 Cox-2 inhibition enhances the activity of sunitinib in human renal cell carcinoma xenografts. Sunitinib 42-51 mitochondrially encoded cytochrome c oxidase II Homo sapiens 0-5 23322198-1 2013 BACKGROUND: Sunitinib (Su), a tyrosine kinase inhibitor of VEGFR, is effective at producing tumour response in clear cell renal cell carcinoma (cRCC), but resistance to therapy is inevitable. Sunitinib 12-21 kinase insert domain receptor Homo sapiens 59-64 23322198-1 2013 BACKGROUND: Sunitinib (Su), a tyrosine kinase inhibitor of VEGFR, is effective at producing tumour response in clear cell renal cell carcinoma (cRCC), but resistance to therapy is inevitable. Sunitinib 12-14 kinase insert domain receptor Homo sapiens 59-64 23322198-11 2013 Clinical trials combining Su and COX-2 inhibitors should be considered as a means delaying time to progression on sunitinib in patients with metastatic cRCC. Sunitinib 114-123 mitochondrially encoded cytochrome c oxidase II Homo sapiens 33-38 23679326-14 2013 In our study, we observed second line sunitinib treatment following IFN-alpha to be effective and tolerable. Sunitinib 38-47 interferon alpha 1 Homo sapiens 68-77 23331791-6 2013 However, HIF-1alpha knockdowns demonstrated relative final volumes that were significantly lower than unmodified tumors when both were treated with bevacizumab (35.88 +- 4.24 vs 53.57 +- 6.61, p=0.0544) or sunitinib (12.46 +- 2.59 vs 36.36 +- 4.82, p=0.0024). Sunitinib 206-215 hypoxia inducible factor 1 subunit alpha Homo sapiens 9-19 23269235-6 2013 Here, we demonstrated that sunitinib significantly increased the levels of LC3-II, concomitant with a decrease of p62 in PC12 cells. Sunitinib 27-36 KH RNA binding domain containing, signal transduction associated 1 Rattus norvegicus 114-117 23269235-8 2013 Furthermore, Atg13 knockdown significantly reduced its protein level, which in turn abolished sunitinib-induced autophagy. Sunitinib 94-103 autophagy related 13 Rattus norvegicus 13-18 23269235-9 2013 Moreover, inhibition of autophagy by siRNAs targeting Atg13 or by pharmacological inhibition with ammonium chloride, enhanced both sunitinib-induced apoptosis and anti-proliferation. Sunitinib 131-140 autophagy related 13 Rattus norvegicus 54-59 23269235-10 2013 Thus, sunitinib-induced autophagy is dependent on the suppression of mTORC1 signaling and the formation of ULK1/2-Atg13-FIP200 complexes. Sunitinib 6-15 CREB regulated transcription coactivator 1 Mus musculus 69-75 23269235-10 2013 Thus, sunitinib-induced autophagy is dependent on the suppression of mTORC1 signaling and the formation of ULK1/2-Atg13-FIP200 complexes. Sunitinib 6-15 unc-51 like autophagy activating kinase 1 Rattus norvegicus 107-111 23269235-10 2013 Thus, sunitinib-induced autophagy is dependent on the suppression of mTORC1 signaling and the formation of ULK1/2-Atg13-FIP200 complexes. Sunitinib 6-15 autophagy related 13 Rattus norvegicus 114-119 23329590-13 2013 Based upon the work presented including a 12.5 % discount for pazopanib, sunitinib was extendedly dominated by a combination of pazopanib and IFN-alpha. Sunitinib 73-82 interferon alpha 1 Homo sapiens 142-151 24649132-0 2013 Successful treatment of erythropoietin-producing advanced renal cell carcinoma after targeted therapy using sunitinib: Case report and review of the literature. Sunitinib 108-117 erythropoietin Homo sapiens 24-38 24649132-3 2013 The present study aimed to report the first case of a patient demonstrating a therapeutic effect on EPO-producing advanced RCC, subsequent to targeted pre-surgical sunitinib therapy, with a review of the literature. Sunitinib 164-173 erythropoietin Homo sapiens 100-103 24649132-9 2013 The EPO-producing RCC was successfully treated following targeted presurgical therapy with sunitinib. Sunitinib 91-100 erythropoietin Homo sapiens 4-7 23555683-12 2013 The immunohistochemical analysis of sunitinib-treated metastatic renal cell carcinomas confirmed the correlation between RAB17, LGALS8, and EPCAM and overall survival. Sunitinib 36-45 RAB17, member RAS oncogene family Homo sapiens 121-126 23555683-12 2013 The immunohistochemical analysis of sunitinib-treated metastatic renal cell carcinomas confirmed the correlation between RAB17, LGALS8, and EPCAM and overall survival. Sunitinib 36-45 galectin 8 Homo sapiens 128-134 23555683-12 2013 The immunohistochemical analysis of sunitinib-treated metastatic renal cell carcinomas confirmed the correlation between RAB17, LGALS8, and EPCAM and overall survival. Sunitinib 36-45 epithelial cell adhesion molecule Homo sapiens 140-145 23294714-5 2013 Moreover, SU11248 treatment concentration-dependently decreased cyclin G1 mRNA and protein expressions (P<0.05), in contrast, it time-dependently increased P27(KIP1); mRNA and protein levels (P<0.05). Sunitinib 10-17 cyclin dependent kinase inhibitor 1B Homo sapiens 163-167 23336010-1 2013 Current inhibitors of angiogenesis comprise either therapeutic antibodies (e.g. bevacicumab binding to VEGF-A) or small molecular inhibitors of receptor tyrosin kinases like e.g. sunitinib, which inhibits PDGFR and VEGFR. Sunitinib 179-188 platelet derived growth factor receptor beta Homo sapiens 205-210 23336010-1 2013 Current inhibitors of angiogenesis comprise either therapeutic antibodies (e.g. bevacicumab binding to VEGF-A) or small molecular inhibitors of receptor tyrosin kinases like e.g. sunitinib, which inhibits PDGFR and VEGFR. Sunitinib 179-188 kinase insert domain receptor Homo sapiens 215-220 23294714-5 2013 Moreover, SU11248 treatment concentration-dependently decreased cyclin G1 mRNA and protein expressions (P<0.05), in contrast, it time-dependently increased P27(KIP1); mRNA and protein levels (P<0.05). Sunitinib 10-17 cyclin G1 Homo sapiens 64-73 24640739-7 2013 Currently, a number of VEGFR2 inhibitors are under clinical trials (ramucirumab, cediranib) and several were approved (sunitinib, sorafenib). Sunitinib 119-128 kinase insert domain receptor Homo sapiens 23-29 23294714-6 2013 CONCLUSION: SU11248 may significantly inhibit the proliferation of CNE-2 cells through up-regulating P27(KIP1); and down-regulating cyclin G1. Sunitinib 12-19 cyclin dependent kinase inhibitor 1B Homo sapiens 101-109 23294714-6 2013 CONCLUSION: SU11248 may significantly inhibit the proliferation of CNE-2 cells through up-regulating P27(KIP1); and down-regulating cyclin G1. Sunitinib 12-19 cyclin G1 Homo sapiens 132-141 23294714-5 2013 Moreover, SU11248 treatment concentration-dependently decreased cyclin G1 mRNA and protein expressions (P<0.05), in contrast, it time-dependently increased P27(KIP1); mRNA and protein levels (P<0.05). Sunitinib 10-17 zinc ribbon domain containing 2 Homo sapiens 159-162 22858558-2 2012 Hypertension (HTN), an on-target class effect of vascular endothelial growth factor signaling-pathway inhibitors, has been shown to correlate with clinical outcome in advanced renal cell carcinoma treated with sunitinib. Sunitinib 210-219 vascular endothelial growth factor A Homo sapiens 49-83 22868821-12 2012 Immunohistochemistry, Western blot, and quantitative polymerase chain reaction results showed both VEGFR-2 and PDGFR-beta expression in the control group was higher than that of the sunitinib-treated group. Sunitinib 182-191 kinase insert domain protein receptor Mus musculus 99-106 22700990-7 2012 In multivariable Cox regression, the skin toxicity was significantly associated with longer OS in the sunitinib cohort. Sunitinib 102-111 cytochrome c oxidase subunit 8A Homo sapiens 17-20 22289686-6 2012 Recent results of the AXIS phase 3 trial demonstrated improved efficacy with second-line axitinib compared with sorafenib in patients who progressed on a variety of first-line therapies, including the VEGFr-TKI sunitinib. Sunitinib 211-220 kinase insert domain receptor Homo sapiens 201-206 22959659-6 2012 Positive PTEN was related to good prognosis with sunitinib (PFS, 15.1 vs. 6.5 months; P = .003) and CKs (OS, 13.7 vs. 7.9 months; P = .039). Sunitinib 49-58 phosphatase and tensin homolog Homo sapiens 9-13 23231522-1 2012 AIM: Sunitinib is an orally active multi-targeted tyrosine kinase inhibitor that exerts its antitumor effects primarily through the selective inhibition of VEGF. Sunitinib 5-14 vascular endothelial growth factor A Homo sapiens 156-160 22644070-6 2012 PTEN demonstrated predictive value of response to sunitinib (70.8 vs. 34.1; p = 0.005). Sunitinib 50-59 phosphatase and tensin homolog Homo sapiens 0-4 22644070-7 2012 p21 was associated with a lower response to sunitinib (35.9 vs. 65.4; p = 0.025). Sunitinib 44-53 H3 histone pseudogene 16 Homo sapiens 0-3 22644070-12 2012 PTEN and p21 may be important in predicting response to sunitinib. Sunitinib 56-65 phosphatase and tensin homolog Homo sapiens 0-4 22644070-12 2012 PTEN and p21 may be important in predicting response to sunitinib. Sunitinib 56-65 H3 histone pseudogene 16 Homo sapiens 9-12 23022045-0 2012 An exploratory study of sunitinib in combination with docetaxel and trastuzumab as first-line therapy for HER2-positive metastatic breast cancer. Sunitinib 24-33 erb-b2 receptor tyrosine kinase 2 Homo sapiens 106-110 23022045-2 2012 METHODS: Patients with unresectable, locally recurrent or metastatic human epidermal growth factor receptor 2 (HER2)+ breast cancer received sunitinib plus docetaxel and trastuzumab. Sunitinib 141-150 erb-b2 receptor tyrosine kinase 2 Homo sapiens 75-109 23022045-2 2012 METHODS: Patients with unresectable, locally recurrent or metastatic human epidermal growth factor receptor 2 (HER2)+ breast cancer received sunitinib plus docetaxel and trastuzumab. Sunitinib 141-150 erb-b2 receptor tyrosine kinase 2 Homo sapiens 111-115 23022045-14 2012 CONCLUSIONS: Sunitinib combined with docetaxel and trastuzumab had an acceptable toxicity profile and showed preliminary antitumor activity as first-line treatment for metastatic HER2+ breast cancer. Sunitinib 13-22 erb-b2 receptor tyrosine kinase 2 Homo sapiens 179-183 22868821-12 2012 Immunohistochemistry, Western blot, and quantitative polymerase chain reaction results showed both VEGFR-2 and PDGFR-beta expression in the control group was higher than that of the sunitinib-treated group. Sunitinib 182-191 platelet derived growth factor receptor, beta polypeptide Mus musculus 111-121 23102636-5 2012 RESULTS: The minimum inhibitory concentrations (IC(min)) of sunitinib quenched its primary targets: FLT-3, VEGFR-2, and RET. Sunitinib 60-69 fms related receptor tyrosine kinase 3 Homo sapiens 100-105 23102636-5 2012 RESULTS: The minimum inhibitory concentrations (IC(min)) of sunitinib quenched its primary targets: FLT-3, VEGFR-2, and RET. Sunitinib 60-69 kinase insert domain receptor Homo sapiens 107-114 23158184-4 2012 Autophagy protein 5 silencing diminished the antiproliferative effects of sunitinib and sorafenib by 44% (P < .05) and 41% (P < .05), respectively, in medullary thyroid cancer-1.1 cells and by 43% (P < .01) and 39% (P < .05), respectively, in TT cells. Sunitinib 74-83 autophagy related 5 Homo sapiens 0-19 23102636-5 2012 RESULTS: The minimum inhibitory concentrations (IC(min)) of sunitinib quenched its primary targets: FLT-3, VEGFR-2, and RET. Sunitinib 60-69 ret proto-oncogene Homo sapiens 120-123 22869928-8 2012 In particular, phospholipase C-gamma1 phosphorylation in sunitinib-resistant tumors was up-regulated by 2.6-fold compared with that in sunitinib-sensitive tumors (p<0.05). Sunitinib 57-66 phospholipase C, gamma 1 Mus musculus 15-37 23047550-10 2012 The PDGFRbeta inhibitors gambogic acid, sunitinib, and tandutinib equally impaired the migration of meningioma cells. Sunitinib 40-49 platelet derived growth factor receptor beta Homo sapiens 4-13 22869928-8 2012 In particular, phospholipase C-gamma1 phosphorylation in sunitinib-resistant tumors was up-regulated by 2.6-fold compared with that in sunitinib-sensitive tumors (p<0.05). Sunitinib 135-144 phospholipase C, gamma 1 Mus musculus 15-37 22912364-1 2012 Sunitinib is an oral, small molecule multitargeted receptor tyrosine kinase inhibitor with antiangiogenic and antitumor activity that primarily targets vascular endothelial growth factor receptors (VEGFRs). Sunitinib 0-9 kinase insert domain receptor Rattus norvegicus 198-204 22912364-9 2012 Because Ser(40) phosphorylation significantly affects TH activity and is known to be regulated by PKC, sunitinib may inhibit Ser(40) phosphorylation via the VEGFR-2/PLC-gamma/PKC pathway. Sunitinib 103-112 kinase insert domain receptor Rattus norvegicus 157-164 22912364-10 2012 Additionally, sunitinib markedly decreased the activity of extracellular signal-regulated kinase (ERK), but not c-Jun NH(2)-terminal kinase or p38 mitogen-activated protein kinase. Sunitinib 14-23 Eph receptor B1 Rattus norvegicus 59-96 22912364-10 2012 Additionally, sunitinib markedly decreased the activity of extracellular signal-regulated kinase (ERK), but not c-Jun NH(2)-terminal kinase or p38 mitogen-activated protein kinase. Sunitinib 14-23 Eph receptor B1 Rattus norvegicus 98-101 22912364-11 2012 Therefore, sunitinib may reduce TH Ser(31) phosphorylation through inhibition of the VEGFR-2/PLC-gamma/PKC/Raf/mitogen-activated protein kinase/extracellular signal-regulated kinase kinase/ERK pathway. Sunitinib 11-20 kinase insert domain receptor Rattus norvegicus 85-92 22912364-11 2012 Therefore, sunitinib may reduce TH Ser(31) phosphorylation through inhibition of the VEGFR-2/PLC-gamma/PKC/Raf/mitogen-activated protein kinase/extracellular signal-regulated kinase kinase/ERK pathway. Sunitinib 11-20 Eph receptor B1 Rattus norvegicus 144-181 22912364-11 2012 Therefore, sunitinib may reduce TH Ser(31) phosphorylation through inhibition of the VEGFR-2/PLC-gamma/PKC/Raf/mitogen-activated protein kinase/extracellular signal-regulated kinase kinase/ERK pathway. Sunitinib 11-20 Eph receptor B1 Rattus norvegicus 189-192 22405734-5 2012 A variety of agents, including sorafenib, sunitinib, cediranib, axitinib, motesanib, linifinib and brivanib inhibit VEGF in addition to either platelet derived growth factor (PDGF), or fibroblast derived growth factor (FGF). Sunitinib 42-51 vascular endothelial growth factor A Homo sapiens 116-120 23036227-2 2012 Small molecule inhibitors such as imatinib and sunitinib are known to inhibit the aberrantly activated KIT and PDGFRA receptor signaling and can lead to excellent clinical outcomes for patients with GIST. Sunitinib 47-56 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 103-106 23036227-2 2012 Small molecule inhibitors such as imatinib and sunitinib are known to inhibit the aberrantly activated KIT and PDGFRA receptor signaling and can lead to excellent clinical outcomes for patients with GIST. Sunitinib 47-56 platelet derived growth factor receptor alpha Homo sapiens 111-117 22955853-5 2012 RESULTS: The presence of EpCAM-positive, live CTCs predicts progression in individual mRCC patient, being associated with distant metastasis under first-line Sunitinib. Sunitinib 158-167 epithelial cell adhesion molecule Homo sapiens 25-30 23032654-11 2012 Although biopsy-proven cases of FSGS associated with sunitinib have been reported, this is, to our knowledge, the first reported case of biopsy-proven FSGS associated with sorafenib. Sunitinib 53-62 actinin alpha 4 Homo sapiens 32-36 22868341-6 2012 The schedule of gemcitabine 800 mg/m(2) on days 1, 8, 15 and sunitinib 25 mg daily was considered to be a MTD. Sunitinib 61-70 metallothionein 1E Homo sapiens 106-109 22948895-0 2012 Polymorphisms in endothelial nitric oxide synthase (eNOS) and vascular endothelial growth factor (VEGF) predict sunitinib-induced hypertension. Sunitinib 112-121 nitric oxide synthase 3 Homo sapiens 17-50 22948895-0 2012 Polymorphisms in endothelial nitric oxide synthase (eNOS) and vascular endothelial growth factor (VEGF) predict sunitinib-induced hypertension. Sunitinib 112-121 nitric oxide synthase 3 Homo sapiens 52-56 22948895-0 2012 Polymorphisms in endothelial nitric oxide synthase (eNOS) and vascular endothelial growth factor (VEGF) predict sunitinib-induced hypertension. Sunitinib 112-121 vascular endothelial growth factor A Homo sapiens 62-96 22948895-0 2012 Polymorphisms in endothelial nitric oxide synthase (eNOS) and vascular endothelial growth factor (VEGF) predict sunitinib-induced hypertension. Sunitinib 112-121 vascular endothelial growth factor A Homo sapiens 98-102 22948895-7 2012 Genetic polymorphisms in VEGFA and eNOS independently predict rise in BP and/or development of severe hypertension in sunitinib-treated patients. Sunitinib 118-127 vascular endothelial growth factor A Homo sapiens 25-30 22819259-0 2012 Blockade of NFkappaB activity by Sunitinib increases cell death in Bortezomib-treated endometrial carcinoma cells. Sunitinib 33-42 nuclear factor kappa B subunit 1 Homo sapiens 12-20 22948895-7 2012 Genetic polymorphisms in VEGFA and eNOS independently predict rise in BP and/or development of severe hypertension in sunitinib-treated patients. Sunitinib 118-127 nitric oxide synthase 3 Homo sapiens 35-39 22327313-3 2012 Therapies targeting the VEGF pathway include bevacizumab, sorafenib, sunitinib, pazopanib, and axitinib, whereas temsirolimus and everolimus inhibit the mTOR pathway. Sunitinib 69-78 vascular endothelial growth factor A Homo sapiens 24-28 22837187-17 2012 Sunitinib serum levels might have been profoundly reduced by mitotane induced cytochrome P450-3A4 activity attenuating its antitumor activity and adverse effects. Sunitinib 0-9 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 78-97 22744707-9 2012 PTC cell lines expressing PDGFR-alpha responded to PDGF-BB stimulation with increased invasive potential and this process can be blocked by the tyrosine kinase receptor inhibitor sunitinib (p < 0.009). Sunitinib 179-188 platelet derived growth factor receptor alpha Homo sapiens 26-37 22671963-0 2012 C-reactive protein as a prognostic marker for advanced renal cell carcinoma treated with sunitinib. Sunitinib 89-98 C-reactive protein Homo sapiens 0-18 22671963-1 2012 OBJECTIVES: To investigate the prognostic role of C-reactive protein in patients with advanced renal cell carcinoma treated with sunitinib. Sunitinib 129-138 C-reactive protein Homo sapiens 50-68 22671963-8 2012 CONCLUSIONS: C-reactive protein is an independent prognostic indicator for patients with advanced renal cell carcinoma treated with sunitinib. Sunitinib 132-141 C-reactive protein Homo sapiens 13-31 22819259-3 2012 In the present study, we demonstrate that the tyrosine kinase receptor inhibitor Sunitinib reduces cell viability, proliferation, clonogenicity and induces apoptotic cell death in endometrial carcinoma cell lines, which is not due to its action through the most known targets like VEGFR, nor through EGFR as demonstrated in this work. Sunitinib 81-90 kinase insert domain receptor Homo sapiens 281-286 22819259-3 2012 In the present study, we demonstrate that the tyrosine kinase receptor inhibitor Sunitinib reduces cell viability, proliferation, clonogenicity and induces apoptotic cell death in endometrial carcinoma cell lines, which is not due to its action through the most known targets like VEGFR, nor through EGFR as demonstrated in this work. Sunitinib 81-90 epidermal growth factor receptor Homo sapiens 282-286 22819259-4 2012 Interestingly, Sunitinib reduces NFkappaB transcriptional activity either at basal level or activation by EGF or TNF-alpha. Sunitinib 15-24 nuclear factor kappa B subunit 1 Homo sapiens 33-41 22819259-4 2012 Interestingly, Sunitinib reduces NFkappaB transcriptional activity either at basal level or activation by EGF or TNF-alpha. Sunitinib 15-24 epidermal growth factor Homo sapiens 106-109 22819259-4 2012 Interestingly, Sunitinib reduces NFkappaB transcriptional activity either at basal level or activation by EGF or TNF-alpha. Sunitinib 15-24 tumor necrosis factor Homo sapiens 113-122 22819259-5 2012 We observed that Sunitinib was able to inhibit the Bortezomib-induced NFkappaB transcriptional activity which correlates with a decrease of the phosphorylated levels of IKKalpha and beta, p65 and IkappaBalpha. Sunitinib 17-26 nuclear factor kappa B subunit 1 Homo sapiens 70-78 22819259-5 2012 We observed that Sunitinib was able to inhibit the Bortezomib-induced NFkappaB transcriptional activity which correlates with a decrease of the phosphorylated levels of IKKalpha and beta, p65 and IkappaBalpha. Sunitinib 17-26 component of inhibitor of nuclear factor kappa B kinase complex Homo sapiens 169-186 22819259-5 2012 We observed that Sunitinib was able to inhibit the Bortezomib-induced NFkappaB transcriptional activity which correlates with a decrease of the phosphorylated levels of IKKalpha and beta, p65 and IkappaBalpha. Sunitinib 17-26 RELA proto-oncogene, NF-kB subunit Homo sapiens 188-191 22819259-5 2012 We observed that Sunitinib was able to inhibit the Bortezomib-induced NFkappaB transcriptional activity which correlates with a decrease of the phosphorylated levels of IKKalpha and beta, p65 and IkappaBalpha. Sunitinib 17-26 NFKB inhibitor alpha Homo sapiens 196-208 23275777-9 2012 The patient"s genotype was wild type for ABCG2 421C>A, which is associated with increased sunitinib exposure. Sunitinib 93-102 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 41-46 23073628-8 2012 Also other clinically used multiselective kinase inhibitors, for instance, sorafenib and sunitinib, have c-Kit included in their range of targets. Sunitinib 89-98 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 105-110 22967802-3 2012 We have investigated the radiosensitizing effects of sunitinib, a potent, multi-tyrosine kinase inhibitor of the VEGFR and PDGFR receptors, on human prostate cancer cells. Sunitinib 53-62 kinase insert domain receptor Homo sapiens 113-118 22967802-3 2012 We have investigated the radiosensitizing effects of sunitinib, a potent, multi-tyrosine kinase inhibitor of the VEGFR and PDGFR receptors, on human prostate cancer cells. Sunitinib 53-62 platelet derived growth factor receptor beta Homo sapiens 123-128 22967802-4 2012 METHODS: The radiosensitizing effects of sunitinib were assessed on human prostate cancer cell lines DU145, PC3 and LNCaP by clonogenic assay. Sunitinib 41-50 chromobox 8 Homo sapiens 108-111 22377563-3 2012 MATERIAL AND METHODS: The primary end point of this randomized phase II trial was the objective response rate according to RECIST criteria and/or Gynecologic Cancer InterGroup CA125 response criteria to sunitinib in patients with recurrent platinum-resistant ovarian cancer who were pretreated with up to three chemotherapies. Sunitinib 203-212 mucin 16, cell surface associated Homo sapiens 176-181 22993368-0 2012 Rapid relapse after resection of a sunitinib-resistant gastrointestinal stromal tumor harboring a secondary mutation in exon 13 of the c-KIT gene. Sunitinib 35-44 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 135-140 22733151-10 2012 In vitro cell line study by immunowestern blotting found that sunitinib malate had an inhibitory effect on the PDGFA pathway by decreasing p-PDGFRA, AKT, and p-AKT expression. Sunitinib 62-78 platelet derived growth factor subunit A Homo sapiens 111-116 22733151-10 2012 In vitro cell line study by immunowestern blotting found that sunitinib malate had an inhibitory effect on the PDGFA pathway by decreasing p-PDGFRA, AKT, and p-AKT expression. Sunitinib 62-78 platelet derived growth factor receptor alpha Homo sapiens 141-147 22733151-10 2012 In vitro cell line study by immunowestern blotting found that sunitinib malate had an inhibitory effect on the PDGFA pathway by decreasing p-PDGFRA, AKT, and p-AKT expression. Sunitinib 62-78 AKT serine/threonine kinase 1 Homo sapiens 149-152 22733151-10 2012 In vitro cell line study by immunowestern blotting found that sunitinib malate had an inhibitory effect on the PDGFA pathway by decreasing p-PDGFRA, AKT, and p-AKT expression. Sunitinib 62-78 AKT serine/threonine kinase 1 Homo sapiens 160-163 22967802-7 2012 RESULTS: Clonogenic survival curve assays for both DU145 and PC3 cells showed that the surviving fraction at 2 Gy was reduced from 0.70 and 0.52 in controls to 0.44 and 0.38, respectively, by a 24 hr pretreatment with 100 nM sunitinib. Sunitinib 225-234 chromobox 8 Homo sapiens 61-64 22967802-9 2012 Dose dependent decreases in VEGFR and PDGFR activation were also observed following sunitinib in both DU145 and PC3 cells. Sunitinib 84-93 kinase insert domain receptor Homo sapiens 28-33 22967802-9 2012 Dose dependent decreases in VEGFR and PDGFR activation were also observed following sunitinib in both DU145 and PC3 cells. Sunitinib 84-93 chromobox 8 Homo sapiens 112-115 22967802-10 2012 We assessed the ability of sunitinib to radiosensitize PC3 xenograft tumors growing in the hind limb of nude mice. Sunitinib 27-36 chromobox 8 Mus musculus 55-58 22317769-4 2012 We hypothesized that administration of sunitinib, a VEGFR inhibitor, following TAE would extend progression-free survival (PFS). Sunitinib 39-48 kinase insert domain receptor Homo sapiens 52-57 22317769-14 2012 Sunitinib, an oral VEGFR inhibitor, can be safely administered following embolization. Sunitinib 0-9 kinase insert domain receptor Homo sapiens 19-24 20884251-9 2012 Bcl-2 apoptotic pathway induction by BCG and sunitinib treatment was evaluated by Western blotting method. Sunitinib 45-54 BCL2 apoptosis regulator Homo sapiens 0-5 22515521-3 2012 Sunitinib (targeting vascular endothelial growth factor), temsirolimus (an inhibitor of the mammalian target of rapamycin - mTOR) and pazopanib (a multi-targeted receptor tyrosine kinase inhibitor) are used in the first line of recurrent disease. Sunitinib 0-9 vascular endothelial growth factor A Homo sapiens 21-55 22970046-7 2012 Sunitinib has been reported to inhibit platelet-derived growth factor receptor (PDGFR) phosphorylation at concentrations over the range of 50-100 ng/ml (sunitinib plus SU12662) in vivo. Sunitinib 0-9 platelet derived growth factor receptor beta Homo sapiens 39-78 22970046-7 2012 Sunitinib has been reported to inhibit platelet-derived growth factor receptor (PDGFR) phosphorylation at concentrations over the range of 50-100 ng/ml (sunitinib plus SU12662) in vivo. Sunitinib 0-9 platelet derived growth factor receptor beta Homo sapiens 80-85 22970046-7 2012 Sunitinib has been reported to inhibit platelet-derived growth factor receptor (PDGFR) phosphorylation at concentrations over the range of 50-100 ng/ml (sunitinib plus SU12662) in vivo. Sunitinib 153-162 platelet derived growth factor receptor beta Homo sapiens 39-78 22970046-7 2012 Sunitinib has been reported to inhibit platelet-derived growth factor receptor (PDGFR) phosphorylation at concentrations over the range of 50-100 ng/ml (sunitinib plus SU12662) in vivo. Sunitinib 153-162 platelet derived growth factor receptor beta Homo sapiens 80-85 22796529-3 2012 Use of 2 distinct approaches resulted in clinical efficacy in blocking the VEGF pathway: small molecule tyrosine kinase inhibitors (sunitinib, sorafenib, axitinib, pazopanib) and the humanized anti-VEGF monoclonal antibody bevacizumab that binds circulating VEGF and prevents activation of the VEGF receptor. Sunitinib 132-141 vascular endothelial growth factor A Homo sapiens 75-79 22129368-0 2012 Endothelial follicle stimulating hormone receptor in primary kidney cancer correlates with subsequent response to sunitinib. Sunitinib 114-123 follicle stimulating hormone receptor Homo sapiens 12-49 22129368-10 2012 The data suggest that FSHR expression levels in the blood vessels of CCRCC primary tumours can be used to predict, with high sensitivity and specificity, the patients who will respond to sunitinib therapy. Sunitinib 187-196 follicle stimulating hormone receptor Homo sapiens 22-26 21735145-5 2012 The mammalian target of rapamycin (mTOR) inhibitor everolimus was approved for use in patients who failed treatment with inhibitors of vascular endothelial growth factor (VEGF) and VEGF receptors (VEGFR) based on the results from a Phase III placebo-controlled study; however, until then, the only licensed agents across the spectrum of mRCC were VEGF(R) inhibitors (sorafenib, sunitinib and bevacizumab + interferon), and as such, a large body of evidence has accumulated regarding their use in sequence. Sunitinib 378-387 mechanistic target of rapamycin kinase Homo sapiens 4-33 21735145-5 2012 The mammalian target of rapamycin (mTOR) inhibitor everolimus was approved for use in patients who failed treatment with inhibitors of vascular endothelial growth factor (VEGF) and VEGF receptors (VEGFR) based on the results from a Phase III placebo-controlled study; however, until then, the only licensed agents across the spectrum of mRCC were VEGF(R) inhibitors (sorafenib, sunitinib and bevacizumab + interferon), and as such, a large body of evidence has accumulated regarding their use in sequence. Sunitinib 378-387 mechanistic target of rapamycin kinase Homo sapiens 35-39 22923165-3 2012 Recently, data from two large placebo-controlled phase III trials have demonstrated that targeted therapies directed against receptor of vascular endothelial growth factor (sunitinib) and mammalian target of rapamycin (mTOR) (everolimus) produced clinically significant improvement in patients with advanced PNETs, resulting in a doubling of progression free survival and leading to their FDA approval. Sunitinib 173-182 vascular endothelial growth factor A Homo sapiens 137-171 22676953-6 2012 The effects of sunitinib malate on angiogenesis and cellular proliferation were measured by immunostaining of CD31 and Ki-67. Sunitinib 15-31 platelet/endothelial cell adhesion molecule 1 Mus musculus 110-114 22676953-10 2012 On targeted contrast-enhanced micro-ultrasound imaging, in vivo vascular endothelial growth factor receptor-2 expression was reduced in the sunitinib group and correlated with a decrease in microvessel density. Sunitinib 140-149 kinase insert domain protein receptor Mus musculus 64-109 20884251-15 2012 Furthermore, the bcl-2 expression was significant reduced in T24 cells in metachronous BCG and sunitinib combination treatment than single agent. Sunitinib 95-104 BCL2 apoptosis regulator Homo sapiens 17-22 22897944-0 2012 Soluble KIT correlates with clinical outcome in patients with metastatic breast cancer treated with sunitinib. Sunitinib 100-109 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 8-11 22266184-0 2012 Sunitinib synergizes the antitumor effect of cisplatin via modulation of ERCC1 expression in models of gastric cancer. Sunitinib 0-9 ERCC excision repair 1, endonuclease non-catalytic subunit Homo sapiens 73-78 22266184-2 2012 Sunitinib showed antiproliferative effect in gastric cancer cells line with high PDGFRA expression. Sunitinib 0-9 platelet derived growth factor receptor alpha Homo sapiens 81-87 22266184-3 2012 Knockdown of PDGFRA showed that sunitinib sensitivity was correlated with the basal expression of PDGFRA. Sunitinib 32-41 platelet derived growth factor receptor alpha Homo sapiens 13-19 22266184-3 2012 Knockdown of PDGFRA showed that sunitinib sensitivity was correlated with the basal expression of PDGFRA. Sunitinib 32-41 platelet derived growth factor receptor alpha Homo sapiens 98-104 22266184-6 2012 We found that sunitinib treatment resulted in the down-regulation of ERCC1 expression via the modulation of PDGFRA expression in gastric cancer cells. Sunitinib 14-23 ERCC excision repair 1, endonuclease non-catalytic subunit Homo sapiens 69-74 22266184-6 2012 We found that sunitinib treatment resulted in the down-regulation of ERCC1 expression via the modulation of PDGFRA expression in gastric cancer cells. Sunitinib 14-23 platelet derived growth factor receptor alpha Homo sapiens 108-114 21953673-2 2012 We here investigated whether inhibition of VEGFR signalin by sunitinib causes changes in plasma proteins associated with tumor endothelium. Sunitinib 61-70 kinase insert domain receptor Homo sapiens 43-48 22897944-1 2012 BACKGROUND: Sunitinib inhibits vascular endothelial growth factor receptors (VEGFRs), platelet-derived growth factor receptors, and stem cell factor receptor (KIT). Sunitinib 12-21 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 159-162 21953673-9 2012 During sunitinib treatment, circulating Ang-2 and sTie-2 significantly decreased (p < 0.001 for both), plasma levels of sVCAM-1 and VEGF significantly increased (p = 0.022 and p < 0.001), whereas those of sICAM-1 and vWF remained stable. Sunitinib 7-16 angiopoietin 2 Homo sapiens 40-45 21953673-9 2012 During sunitinib treatment, circulating Ang-2 and sTie-2 significantly decreased (p < 0.001 for both), plasma levels of sVCAM-1 and VEGF significantly increased (p = 0.022 and p < 0.001), whereas those of sICAM-1 and vWF remained stable. Sunitinib 7-16 vascular endothelial growth factor A Homo sapiens 135-139 22897944-10 2012 CONCLUSIONS: This exploratory analysis suggests that changes in sKIT and possibly VEGF-A early during sunitinib treatment may be predictive of clinical outcome in MBC. Sunitinib 102-111 vascular endothelial growth factor A Homo sapiens 82-88 21953673-9 2012 During sunitinib treatment, circulating Ang-2 and sTie-2 significantly decreased (p < 0.001 for both), plasma levels of sVCAM-1 and VEGF significantly increased (p = 0.022 and p < 0.001), whereas those of sICAM-1 and vWF remained stable. Sunitinib 7-16 von Willebrand factor Homo sapiens 223-226 21953673-13 2012 In conclusion, sunitinib-induced changes in Ang-2, sTie-2, sVCAM-1 and VEGF are related to the administration schedule, while reduction in Ang-2 is also associated with decrease in tumor burden. Sunitinib 15-24 angiopoietin 2 Homo sapiens 44-49 22391574-0 2012 Sunitinib targets PDGF-receptor and Flt3 and reduces survival and migration of human meningioma cells. Sunitinib 0-9 fms related receptor tyrosine kinase 3 Homo sapiens 36-40 21953673-13 2012 In conclusion, sunitinib-induced changes in Ang-2, sTie-2, sVCAM-1 and VEGF are related to the administration schedule, while reduction in Ang-2 is also associated with decrease in tumor burden. Sunitinib 15-24 vascular endothelial growth factor A Homo sapiens 71-75 21953673-13 2012 In conclusion, sunitinib-induced changes in Ang-2, sTie-2, sVCAM-1 and VEGF are related to the administration schedule, while reduction in Ang-2 is also associated with decrease in tumor burden. Sunitinib 15-24 angiopoietin 2 Homo sapiens 139-144 22207048-8 2012 The effect of VEGF, bevacizumab, and sunitinib on the phosphorylation of VEGFR-2 and downstream effecter molecules, MAPK and PI3K, was examined by western blot. Sunitinib 37-46 kinase insert domain receptor Homo sapiens 73-80 22207048-8 2012 The effect of VEGF, bevacizumab, and sunitinib on the phosphorylation of VEGFR-2 and downstream effecter molecules, MAPK and PI3K, was examined by western blot. Sunitinib 37-46 mitogen-activated protein kinase 1 Homo sapiens 116-120 22207048-12 2012 VEGF/VEGFR inhibitors, bevacizumab and sunitinib, significantly decreased the motility of pancreas cancer cells. Sunitinib 39-48 vascular endothelial growth factor A Homo sapiens 0-4 22207048-12 2012 VEGF/VEGFR inhibitors, bevacizumab and sunitinib, significantly decreased the motility of pancreas cancer cells. Sunitinib 39-48 kinase insert domain receptor Homo sapiens 5-10 22207048-14 2012 Bevacizumab and sunitinib decreased the level of VEGFR-2 phosphorylation, p-ERK, and p-Akt expression. Sunitinib 16-25 kinase insert domain receptor Homo sapiens 49-56 22207048-14 2012 Bevacizumab and sunitinib decreased the level of VEGFR-2 phosphorylation, p-ERK, and p-Akt expression. Sunitinib 16-25 mitogen-activated protein kinase 1 Homo sapiens 76-79 22532265-2 2012 Multiple VEGF inhibiting orally administered tyrosine kinase inhibitors (TKIs) have been approved including sunitinib, sorafenib, pazopanib and most recently, axitinib. Sunitinib 108-117 vascular endothelial growth factor A Homo sapiens 9-13 22336056-1 2012 BACKGROUND: This exploratory study examined the pharmacokinetics, safety, and antitumor activity of sunitinib plus docetaxel in patients with HER-2-negative advanced breast cancer. Sunitinib 100-109 erb-b2 receptor tyrosine kinase 2 Homo sapiens 142-147 22391574-9 2012 Western blot analyses showed abolished platelet derived growth factor receptor (PDGFR)-autophosphorylation after sunitinib. Sunitinib 113-122 platelet derived growth factor receptor beta Homo sapiens 39-78 22391574-9 2012 Western blot analyses showed abolished platelet derived growth factor receptor (PDGFR)-autophosphorylation after sunitinib. Sunitinib 113-122 platelet derived growth factor receptor beta Homo sapiens 80-85 22572725-3 2012 RECENT FINDINGS: Recent studies with oral vascular endothelial growth factor receptor (VEGFR) inhibitors such as sunitinib, sorafenib and cediranib have shown disease stabilization in patients with advanced synovial sarcoma. Sunitinib 113-122 kinase insert domain receptor Homo sapiens 42-85 22611036-7 2012 Consistent with these results, sunitinib treatment resulted in an up-regulation of the hypoxia marker GLUT1 in PNETs, whereas anti-VEGF did not. Sunitinib 31-40 solute carrier family 2 (facilitated glucose transporter), member 1 Mus musculus 102-107 22665524-7 2012 Notably our data indicate that sorafenib is more effective than imatinib or sunitinib for inhibiting the kinase activity of drug-resistant KIT mutants (as assessed by biochemical IC(50)). Sunitinib 76-85 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 139-142 22463589-10 2012 Moreover, Fuhrman grading and concomitant, CXCR4 and Fuhrman grading, strongly predicted sunitinib first line therapy responsiveness on progression-free survival and overall survival. Sunitinib 89-98 C-X-C motif chemokine receptor 4 Homo sapiens 43-48 22463589-11 2012 CONCLUSIONS: High CXCR4 expression correlates with sunitinib poor response in metastatic renal cancer. Sunitinib 51-60 C-X-C motif chemokine receptor 4 Homo sapiens 18-23 22560921-2 2012 Sunitinib, an oral agent that inhibits the VEGF signaling pathway, may delay progression in sequence with chemotherapy. Sunitinib 0-9 vascular endothelial growth factor A Homo sapiens 43-47 22020623-0 2012 PRKX, TTBK2 and RSK4 expression causes Sunitinib resistance in kidney carcinoma- and melanoma-cell lines. Sunitinib 39-48 protein kinase X-linked Homo sapiens 0-4 22020623-0 2012 PRKX, TTBK2 and RSK4 expression causes Sunitinib resistance in kidney carcinoma- and melanoma-cell lines. Sunitinib 39-48 tau tubulin kinase 2 Homo sapiens 6-11 22020623-0 2012 PRKX, TTBK2 and RSK4 expression causes Sunitinib resistance in kidney carcinoma- and melanoma-cell lines. Sunitinib 39-48 ribosomal protein S6 kinase A6 Homo sapiens 16-20 22020623-7 2012 Therefore, development of specific or multi-targeted inhibitors for these kinases for combinatorial therapy with e.g., an IL-8 neutralizing antibody might circumvent or substantially delay Sunitinib resistance formation and enhance survival prognosis. Sunitinib 189-198 C-X-C motif chemokine ligand 8 Homo sapiens 122-126 22020623-10 2012 In line with the elevated expression of PRKX, TTBK2 or RSK4, this sensitization effect was strikingly higher in the Sunitinib resistant cell lines, suggesting an expression-based mechanism of these genes to trigger Sunitinib resistance. Sunitinib 116-125 protein kinase X-linked Homo sapiens 40-44 22020623-10 2012 In line with the elevated expression of PRKX, TTBK2 or RSK4, this sensitization effect was strikingly higher in the Sunitinib resistant cell lines, suggesting an expression-based mechanism of these genes to trigger Sunitinib resistance. Sunitinib 116-125 tau tubulin kinase 2 Homo sapiens 46-51 22020623-10 2012 In line with the elevated expression of PRKX, TTBK2 or RSK4, this sensitization effect was strikingly higher in the Sunitinib resistant cell lines, suggesting an expression-based mechanism of these genes to trigger Sunitinib resistance. Sunitinib 116-125 ribosomal protein S6 kinase A6 Homo sapiens 55-59 22020623-10 2012 In line with the elevated expression of PRKX, TTBK2 or RSK4, this sensitization effect was strikingly higher in the Sunitinib resistant cell lines, suggesting an expression-based mechanism of these genes to trigger Sunitinib resistance. Sunitinib 215-224 protein kinase X-linked Homo sapiens 40-44 22020623-10 2012 In line with the elevated expression of PRKX, TTBK2 or RSK4, this sensitization effect was strikingly higher in the Sunitinib resistant cell lines, suggesting an expression-based mechanism of these genes to trigger Sunitinib resistance. Sunitinib 215-224 tau tubulin kinase 2 Homo sapiens 46-51 22020623-10 2012 In line with the elevated expression of PRKX, TTBK2 or RSK4, this sensitization effect was strikingly higher in the Sunitinib resistant cell lines, suggesting an expression-based mechanism of these genes to trigger Sunitinib resistance. Sunitinib 215-224 ribosomal protein S6 kinase A6 Homo sapiens 55-59 22020623-11 2012 Hence, we propose that PRKX, TTBK2 and RSK4 are potential resistance markers in Sunitinib therapy and might therefore represent targets for the development of novel strategies to overcome resistance. Sunitinib 80-89 protein kinase X-linked Homo sapiens 23-27 22020623-11 2012 Hence, we propose that PRKX, TTBK2 and RSK4 are potential resistance markers in Sunitinib therapy and might therefore represent targets for the development of novel strategies to overcome resistance. Sunitinib 80-89 tau tubulin kinase 2 Homo sapiens 29-34 22020623-11 2012 Hence, we propose that PRKX, TTBK2 and RSK4 are potential resistance markers in Sunitinib therapy and might therefore represent targets for the development of novel strategies to overcome resistance. Sunitinib 80-89 ribosomal protein S6 kinase A6 Homo sapiens 39-43 22269210-0 2012 HGF/c-Met pathway is one of the mediators of sunitinib-induced tumor cell type-dependent metastasis. Sunitinib 45-54 hepatocyte growth factor Mus musculus 0-3 22269210-0 2012 HGF/c-Met pathway is one of the mediators of sunitinib-induced tumor cell type-dependent metastasis. Sunitinib 45-54 met proto-oncogene Mus musculus 4-9 22269210-7 2012 In conclusion, our findings indicate that sunitinib-induced metastasis is tumor cell-type dependent and further supports a rationale for combination of anti-angiogenics and c-Met inhibition in the clinic. Sunitinib 42-51 met proto-oncogene Mus musculus 173-178 22463589-0 2012 High CXCR4 expression correlates with sunitinib poor response in metastatic renal cancer. Sunitinib 38-47 C-X-C motif chemokine receptor 4 Homo sapiens 5-10 22463589-3 2012 It was hypothesized that CXCR4 expression in primary renal cancer could predict sunitinib responsiveness. Sunitinib 80-89 C-X-C motif chemokine receptor 4 Homo sapiens 25-30 22463589-9 2012 Low or absent CXCR4 expression predicted response to sunitinib therapy. Sunitinib 53-62 C-X-C motif chemokine receptor 4 Homo sapiens 14-19 22572725-3 2012 RECENT FINDINGS: Recent studies with oral vascular endothelial growth factor receptor (VEGFR) inhibitors such as sunitinib, sorafenib and cediranib have shown disease stabilization in patients with advanced synovial sarcoma. Sunitinib 113-122 kinase insert domain receptor Homo sapiens 87-92 22213155-0 2012 Sunitinib inhibits the phosphorylation of platelet-derived growth factor receptor beta in the skin of mice with scleroderma-like features and prevents the development of the disease. Sunitinib 0-9 platelet derived growth factor receptor, beta polypeptide Mus musculus 42-86 23264850-6 2012 Pimozide shows a combinatorial effect with the tyrosine kinase inhibitors midostaurin (PKC412) and sunitinib in the inhibition of STAT5 tyrosine phosphorylation and the induction of apoptosis. Sunitinib 99-108 signal transducer and activator of transcription 5A Mus musculus 130-135 22904678-6 2012 Small-molecule tyrosine kinase inhibitor, sunitinib, caused an inhibition of VEGFR2 phosphorylation in WM239 but not in WM115 cells. Sunitinib 42-51 kinase insert domain receptor Homo sapiens 77-83 22080184-2 2012 This study aimed to assess the antitumor activity of sunitinib, a multi-targeted inhibitor of vascular endothelial growth factor receptor, c-kit, platelet-derived growth factor receptor, ret proto-oncogene (RET) and FMS-like tyrosine kinase 3 (FLT3), in ACC of the salivary gland. Sunitinib 53-62 vascular endothelial growth factor A Homo sapiens 94-128 22080184-2 2012 This study aimed to assess the antitumor activity of sunitinib, a multi-targeted inhibitor of vascular endothelial growth factor receptor, c-kit, platelet-derived growth factor receptor, ret proto-oncogene (RET) and FMS-like tyrosine kinase 3 (FLT3), in ACC of the salivary gland. Sunitinib 53-62 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 139-144 22080184-2 2012 This study aimed to assess the antitumor activity of sunitinib, a multi-targeted inhibitor of vascular endothelial growth factor receptor, c-kit, platelet-derived growth factor receptor, ret proto-oncogene (RET) and FMS-like tyrosine kinase 3 (FLT3), in ACC of the salivary gland. Sunitinib 53-62 ret proto-oncogene Homo sapiens 187-190 22080184-2 2012 This study aimed to assess the antitumor activity of sunitinib, a multi-targeted inhibitor of vascular endothelial growth factor receptor, c-kit, platelet-derived growth factor receptor, ret proto-oncogene (RET) and FMS-like tyrosine kinase 3 (FLT3), in ACC of the salivary gland. Sunitinib 53-62 ret proto-oncogene Homo sapiens 207-210 22080184-2 2012 This study aimed to assess the antitumor activity of sunitinib, a multi-targeted inhibitor of vascular endothelial growth factor receptor, c-kit, platelet-derived growth factor receptor, ret proto-oncogene (RET) and FMS-like tyrosine kinase 3 (FLT3), in ACC of the salivary gland. Sunitinib 53-62 fms related receptor tyrosine kinase 3 Homo sapiens 216-242 22080184-2 2012 This study aimed to assess the antitumor activity of sunitinib, a multi-targeted inhibitor of vascular endothelial growth factor receptor, c-kit, platelet-derived growth factor receptor, ret proto-oncogene (RET) and FMS-like tyrosine kinase 3 (FLT3), in ACC of the salivary gland. Sunitinib 53-62 fms related receptor tyrosine kinase 3 Homo sapiens 244-248 22532600-0 2012 Modulation of Akt/mTOR signaling overcomes sunitinib resistance in renal and prostate cancer cells. Sunitinib 43-52 AKT serine/threonine kinase 1 Homo sapiens 14-17 22532600-0 2012 Modulation of Akt/mTOR signaling overcomes sunitinib resistance in renal and prostate cancer cells. Sunitinib 43-52 mechanistic target of rapamycin kinase Homo sapiens 18-22 22532600-4 2012 Our experiments showed that PTEN expression inversely correlates with sunitinib resistance in renal and prostate cancer cells. Sunitinib 70-79 phosphatase and tensin homolog Homo sapiens 28-32 22532600-5 2012 Restoration of PTEN expression markedly increases sensitivity of tumor cells to sunitinib both in vitro and in vivo. Sunitinib 80-89 phosphatase and tensin homolog Homo sapiens 15-19 22532600-6 2012 In addition, pharmacologic manipulation of PI3K/Akt/mTOR signaling with PI3K/mTOR inhibitor, GDC-0980, mTOR inhibitor, temsirolimus, or pan-Akt inhibitor, GSK690693, was able to overcome sunitinib resistance in cancer cells. Sunitinib 187-196 mechanistic target of rapamycin kinase Homo sapiens 52-56 22532600-7 2012 Our findings underscore the importance of PTEN expression in relation to sunitinib resistance and imply a direct cytotoxic effect by sunitinib on tumor cells in addition to its antiangiogenic actions. Sunitinib 73-82 phosphatase and tensin homolog Homo sapiens 42-46 21956360-8 2012 Patients presented with a mean elevation of parathyroid hormone after 2.2 cycles of sunitinib. Sunitinib 84-93 parathyroid hormone Homo sapiens 44-63 21956360-11 2012 Parathyroid hormone elevation usually persisted but did not progress during long-term therapy with sunitinib. Sunitinib 99-108 parathyroid hormone Homo sapiens 0-19 22213155-12 2012 Sunitinib significantly reduced the phosphorylation of both PDGF and VEGF receptors. Sunitinib 0-9 vascular endothelial growth factor A Mus musculus 69-73 22213155-13 2012 CONCLUSION: Inhibition of the hyperactivated PDGF and VEGF pathways by sunitinib prevented the development of fibrosis in HOCl-induced murine SSc and may represent a new SSc treatment for testing in clinical trials. Sunitinib 71-80 vascular endothelial growth factor A Mus musculus 54-58 22442268-0 2012 Activation of the RAS/RAF/ERK signaling pathway contributes to resistance to sunitinib in thyroid carcinoma cell lines. Sunitinib 77-86 zinc fingers and homeoboxes 2 Homo sapiens 22-25 22607009-2 2012 Sunitinib is an oral, small-molecule, multi-targeted receptor tyrosine kinase inhibitor (TKI) that targets a number of receptors, including vascular endothelial growth factor receptors (VEGFR) and platelet-derived growth factor receptors (PDGFR). Sunitinib 0-9 kinase insert domain receptor Homo sapiens 140-184 22607009-2 2012 Sunitinib is an oral, small-molecule, multi-targeted receptor tyrosine kinase inhibitor (TKI) that targets a number of receptors, including vascular endothelial growth factor receptors (VEGFR) and platelet-derived growth factor receptors (PDGFR). Sunitinib 0-9 kinase insert domain receptor Homo sapiens 186-191 22442268-4 2012 RESULTS: Sunitinib was found to selectively inhibit cell proliferation, induce cell accumulation in the G0-G1 phase, and inhibit the phosphorylation of ERK1/2 in both KRAS/BRAF wild-type thyroid cancer cells and in tumor cells harboring the RET/PTC rearrangement, whereas it was completely ineffective in KRAS- or BRAF-mutated thyroid carcinoma cells. Sunitinib 9-18 mitogen-activated protein kinase 3 Homo sapiens 152-158 22442268-4 2012 RESULTS: Sunitinib was found to selectively inhibit cell proliferation, induce cell accumulation in the G0-G1 phase, and inhibit the phosphorylation of ERK1/2 in both KRAS/BRAF wild-type thyroid cancer cells and in tumor cells harboring the RET/PTC rearrangement, whereas it was completely ineffective in KRAS- or BRAF-mutated thyroid carcinoma cells. Sunitinib 9-18 KRAS proto-oncogene, GTPase Homo sapiens 167-171 21792888-7 2012 Molecular studies in Caki-1 showed that saracatinib alone and in combination with sunitinib inhibited phosphorylation of the cell progression regulator c-Myc in a dose-dependent manner. Sunitinib 82-91 MYC proto-oncogene, bHLH transcription factor Homo sapiens 152-157 21792888-8 2012 Sunitinib alone or in combination suppressed cyclin-D1 expression with the combination showing greater dose-dependent effect. Sunitinib 0-9 cyclin D1 Homo sapiens 45-54 21792888-9 2012 Sunitinib inhibited vascular endothelial growth factor (VEGF) secretion through the inhibition of STAT3 signaling and VEGF biosynthesis. Sunitinib 0-9 vascular endothelial growth factor A Homo sapiens 56-60 21792888-9 2012 Sunitinib inhibited vascular endothelial growth factor (VEGF) secretion through the inhibition of STAT3 signaling and VEGF biosynthesis. Sunitinib 0-9 signal transducer and activator of transcription 3 Homo sapiens 98-103 21792888-9 2012 Sunitinib inhibited vascular endothelial growth factor (VEGF) secretion through the inhibition of STAT3 signaling and VEGF biosynthesis. Sunitinib 0-9 vascular endothelial growth factor A Homo sapiens 118-122 21792888-10 2012 HIF1-alpha expression in normoxic and hypoxic conditions in Caki-1 cells was inhibited by either saracatinib or sunitinib when administered alone, however, a greater reduction occurred when these compounds were given in combination. Sunitinib 112-121 hypoxia inducible factor 1 subunit alpha Homo sapiens 0-10 22442268-4 2012 RESULTS: Sunitinib was found to selectively inhibit cell proliferation, induce cell accumulation in the G0-G1 phase, and inhibit the phosphorylation of ERK1/2 in both KRAS/BRAF wild-type thyroid cancer cells and in tumor cells harboring the RET/PTC rearrangement, whereas it was completely ineffective in KRAS- or BRAF-mutated thyroid carcinoma cells. Sunitinib 9-18 B-Raf proto-oncogene, serine/threonine kinase Homo sapiens 172-176 22442268-4 2012 RESULTS: Sunitinib was found to selectively inhibit cell proliferation, induce cell accumulation in the G0-G1 phase, and inhibit the phosphorylation of ERK1/2 in both KRAS/BRAF wild-type thyroid cancer cells and in tumor cells harboring the RET/PTC rearrangement, whereas it was completely ineffective in KRAS- or BRAF-mutated thyroid carcinoma cells. Sunitinib 9-18 ret proto-oncogene Homo sapiens 241-244 22442268-4 2012 RESULTS: Sunitinib was found to selectively inhibit cell proliferation, induce cell accumulation in the G0-G1 phase, and inhibit the phosphorylation of ERK1/2 in both KRAS/BRAF wild-type thyroid cancer cells and in tumor cells harboring the RET/PTC rearrangement, whereas it was completely ineffective in KRAS- or BRAF-mutated thyroid carcinoma cells. Sunitinib 9-18 ret proto-oncogene Homo sapiens 245-248 22442268-4 2012 RESULTS: Sunitinib was found to selectively inhibit cell proliferation, induce cell accumulation in the G0-G1 phase, and inhibit the phosphorylation of ERK1/2 in both KRAS/BRAF wild-type thyroid cancer cells and in tumor cells harboring the RET/PTC rearrangement, whereas it was completely ineffective in KRAS- or BRAF-mutated thyroid carcinoma cells. Sunitinib 9-18 KRAS proto-oncogene, GTPase Homo sapiens 305-310 22442268-4 2012 RESULTS: Sunitinib was found to selectively inhibit cell proliferation, induce cell accumulation in the G0-G1 phase, and inhibit the phosphorylation of ERK1/2 in both KRAS/BRAF wild-type thyroid cancer cells and in tumor cells harboring the RET/PTC rearrangement, whereas it was completely ineffective in KRAS- or BRAF-mutated thyroid carcinoma cells. Sunitinib 9-18 B-Raf proto-oncogene, serine/threonine kinase Homo sapiens 314-318 22442268-0 2012 Activation of the RAS/RAF/ERK signaling pathway contributes to resistance to sunitinib in thyroid carcinoma cell lines. Sunitinib 77-86 mitogen-activated protein kinase 1 Homo sapiens 26-29 22442268-6 2012 Of note, the constitutive activation of RAS/RAF/ERK signaling in KRAS/BRAF wild-type cells by transfection of the R12 HRAS or V600E BRAF mutants or stimulation with epithelial growth factor resulted in the loss of responsiveness to sunitinib, whereas pharmacological inhibition of MAPK kinase activity resulted in the resensitization of KRAS- or BRAF-mutated cells to the multikinase inhibitor. Sunitinib 232-241 zinc fingers and homeoboxes 2 Homo sapiens 44-47 22442268-6 2012 Of note, the constitutive activation of RAS/RAF/ERK signaling in KRAS/BRAF wild-type cells by transfection of the R12 HRAS or V600E BRAF mutants or stimulation with epithelial growth factor resulted in the loss of responsiveness to sunitinib, whereas pharmacological inhibition of MAPK kinase activity resulted in the resensitization of KRAS- or BRAF-mutated cells to the multikinase inhibitor. Sunitinib 232-241 mitogen-activated protein kinase 1 Homo sapiens 48-51 22442268-1 2012 CONTEXT: Sunitinib is currently being evaluated in advanced human thyroid carcinomas, based on the rationale that the vascular endothelial growth factor and platelet-derived growth factor receptors and the RET/PTC rearrangement are valuable targets for the treatment of this malignancy. Sunitinib 9-18 vascular endothelial growth factor A Homo sapiens 118-152 22442268-6 2012 Of note, the constitutive activation of RAS/RAF/ERK signaling in KRAS/BRAF wild-type cells by transfection of the R12 HRAS or V600E BRAF mutants or stimulation with epithelial growth factor resulted in the loss of responsiveness to sunitinib, whereas pharmacological inhibition of MAPK kinase activity resulted in the resensitization of KRAS- or BRAF-mutated cells to the multikinase inhibitor. Sunitinib 232-241 KRAS proto-oncogene, GTPase Homo sapiens 65-69 22442268-1 2012 CONTEXT: Sunitinib is currently being evaluated in advanced human thyroid carcinomas, based on the rationale that the vascular endothelial growth factor and platelet-derived growth factor receptors and the RET/PTC rearrangement are valuable targets for the treatment of this malignancy. Sunitinib 9-18 ret proto-oncogene Homo sapiens 206-209 22442268-6 2012 Of note, the constitutive activation of RAS/RAF/ERK signaling in KRAS/BRAF wild-type cells by transfection of the R12 HRAS or V600E BRAF mutants or stimulation with epithelial growth factor resulted in the loss of responsiveness to sunitinib, whereas pharmacological inhibition of MAPK kinase activity resulted in the resensitization of KRAS- or BRAF-mutated cells to the multikinase inhibitor. Sunitinib 232-241 B-Raf proto-oncogene, serine/threonine kinase Homo sapiens 70-74 22442268-6 2012 Of note, the constitutive activation of RAS/RAF/ERK signaling in KRAS/BRAF wild-type cells by transfection of the R12 HRAS or V600E BRAF mutants or stimulation with epithelial growth factor resulted in the loss of responsiveness to sunitinib, whereas pharmacological inhibition of MAPK kinase activity resulted in the resensitization of KRAS- or BRAF-mutated cells to the multikinase inhibitor. Sunitinib 232-241 mitogen-activated protein kinase 3 Homo sapiens 281-285 22442268-1 2012 CONTEXT: Sunitinib is currently being evaluated in advanced human thyroid carcinomas, based on the rationale that the vascular endothelial growth factor and platelet-derived growth factor receptors and the RET/PTC rearrangement are valuable targets for the treatment of this malignancy. Sunitinib 9-18 ret proto-oncogene Homo sapiens 210-213 22442268-6 2012 Of note, the constitutive activation of RAS/RAF/ERK signaling in KRAS/BRAF wild-type cells by transfection of the R12 HRAS or V600E BRAF mutants or stimulation with epithelial growth factor resulted in the loss of responsiveness to sunitinib, whereas pharmacological inhibition of MAPK kinase activity resulted in the resensitization of KRAS- or BRAF-mutated cells to the multikinase inhibitor. Sunitinib 232-241 KRAS proto-oncogene, GTPase Homo sapiens 337-342 22442268-7 2012 CONCLUSIONS: The constitutive activation of the RAS/RAF/ERK pathway may favor resistance to sunitinib in thyroid carcinoma cells. Sunitinib 92-101 zinc fingers and homeoboxes 2 Homo sapiens 52-55 22442268-3 2012 DESIGN: The effect of activating somatic mutations in the KRAS and BRAF genes on the responsiveness to sunitinib was evaluated in a panel of thyroid cancer cell lines harboring wild-type KRAS and BRAF genes, the RET/PTC1 rearrangement, the G12R KRAS, or the V600E BRAF mutation. Sunitinib 103-112 KRAS proto-oncogene, GTPase Homo sapiens 58-62 22442268-7 2012 CONCLUSIONS: The constitutive activation of the RAS/RAF/ERK pathway may favor resistance to sunitinib in thyroid carcinoma cells. Sunitinib 92-101 mitogen-activated protein kinase 1 Homo sapiens 56-59 22442268-3 2012 DESIGN: The effect of activating somatic mutations in the KRAS and BRAF genes on the responsiveness to sunitinib was evaluated in a panel of thyroid cancer cell lines harboring wild-type KRAS and BRAF genes, the RET/PTC1 rearrangement, the G12R KRAS, or the V600E BRAF mutation. Sunitinib 103-112 B-Raf proto-oncogene, serine/threonine kinase Homo sapiens 67-71 22442268-3 2012 DESIGN: The effect of activating somatic mutations in the KRAS and BRAF genes on the responsiveness to sunitinib was evaluated in a panel of thyroid cancer cell lines harboring wild-type KRAS and BRAF genes, the RET/PTC1 rearrangement, the G12R KRAS, or the V600E BRAF mutation. Sunitinib 103-112 B-Raf proto-oncogene, serine/threonine kinase Homo sapiens 196-200 22442268-3 2012 DESIGN: The effect of activating somatic mutations in the KRAS and BRAF genes on the responsiveness to sunitinib was evaluated in a panel of thyroid cancer cell lines harboring wild-type KRAS and BRAF genes, the RET/PTC1 rearrangement, the G12R KRAS, or the V600E BRAF mutation. Sunitinib 103-112 ret proto-oncogene Homo sapiens 212-215 22442268-3 2012 DESIGN: The effect of activating somatic mutations in the KRAS and BRAF genes on the responsiveness to sunitinib was evaluated in a panel of thyroid cancer cell lines harboring wild-type KRAS and BRAF genes, the RET/PTC1 rearrangement, the G12R KRAS, or the V600E BRAF mutation. Sunitinib 103-112 patched 1 Homo sapiens 216-220 22442268-3 2012 DESIGN: The effect of activating somatic mutations in the KRAS and BRAF genes on the responsiveness to sunitinib was evaluated in a panel of thyroid cancer cell lines harboring wild-type KRAS and BRAF genes, the RET/PTC1 rearrangement, the G12R KRAS, or the V600E BRAF mutation. Sunitinib 103-112 B-Raf proto-oncogene, serine/threonine kinase Homo sapiens 196-200 22932406-10 2012 CONCLUSION: The c-kit/PDGFRalpha genotypes after imatinib mesylate resistance may both relate to primary mutations and efficacy of sunitinib treatment. Sunitinib 131-140 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 16-21 22579211-0 2012 Re: Targeting PIM kinase enhances the activity of sunitinib in renal cell carcinoma. Sunitinib 50-59 Pim-1 proto-oncogene, serine/threonine kinase Homo sapiens 14-17 21455800-9 2012 Both docetaxel and sunitinib malate had no effect on each other in inhibiting ERK1/2 phosphorylation in sequential treatments, while docetaxel eliminated inhibitory activity of sunitinib malate on ERK1/2 phosphorylation in concurrent treatment. Sunitinib 177-193 mitogen-activated protein kinase 3 Homo sapiens 197-203 21455800-11 2012 Combined treatment with sunitinib malate and docetaxel had a greater therapeutic effect than monotherapy, and the first sequential scheduling was more effective than concurrent scheduling, which partly due to the effect of docetaxel on receptor tyrosine kinase (RTK) signaling pathway. Sunitinib 24-40 ret proto-oncogene Homo sapiens 236-260 21455800-11 2012 Combined treatment with sunitinib malate and docetaxel had a greater therapeutic effect than monotherapy, and the first sequential scheduling was more effective than concurrent scheduling, which partly due to the effect of docetaxel on receptor tyrosine kinase (RTK) signaling pathway. Sunitinib 24-40 ret proto-oncogene Homo sapiens 262-265 22783417-8 2012 There was a tendency for a positive correlation between the sensitivity to sunitinib and platelet-derived growth factor beta (PDGFR-beta) expression levels, which were examined by reverse transcription polymerase chain reaction. Sunitinib 75-84 platelet derived growth factor receptor beta Homo sapiens 126-136 22585430-3 2012 When exposed to vascular endothelial growth factor (VEGFR) inhibitors such as sunitinib, target lesions display few if any variation in tumor size, but rather detectable modifications in tumor density. Sunitinib 78-87 vascular endothelial growth factor A Homo sapiens 16-50 22585430-3 2012 When exposed to vascular endothelial growth factor (VEGFR) inhibitors such as sunitinib, target lesions display few if any variation in tumor size, but rather detectable modifications in tumor density. Sunitinib 78-87 vascular endothelial growth factor A Homo sapiens 52-57 22585430-7 2012 Choi criteria have been recently proposed as a surrogate endpoint for efficacy and to identify patients that are good responders to VEGFR inhibitors such as sunitinib and sorafenib in advanced hepatocellular carcinoma, another disease highly addicted to angiogenesis. Sunitinib 157-166 vascular endothelial growth factor A Homo sapiens 132-137 22592949-3 2012 Therefore, several antiangiogenic compounds have been tested in these malignancies, and among these, sunitinib has demonstrated activity in pancreatic NET patients by dually targeting the VEGFR and PDGFR pathways. Sunitinib 101-110 kinase insert domain receptor Homo sapiens 188-193 22592949-3 2012 Therefore, several antiangiogenic compounds have been tested in these malignancies, and among these, sunitinib has demonstrated activity in pancreatic NET patients by dually targeting the VEGFR and PDGFR pathways. Sunitinib 101-110 platelet derived growth factor receptor beta Homo sapiens 198-203 22932406-0 2012 [Secondary mutation of c-kit/PDGFRalpha genotypes after imatinib mesylate therapy and its relationship with efficacy of sunitinib]. Sunitinib 120-129 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 23-28 22932406-0 2012 [Secondary mutation of c-kit/PDGFRalpha genotypes after imatinib mesylate therapy and its relationship with efficacy of sunitinib]. Sunitinib 120-129 platelet derived growth factor receptor alpha Homo sapiens 29-39 22932406-1 2012 OBJECTIVE: To investigate the relationship between secondary mutations of c-kit/PDGFRalpha resistance to imatinib mesylate and the efficacy of sunitinib in patients with gastrointestinal stromal tumor (GIST). Sunitinib 143-152 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 74-79 22932406-10 2012 CONCLUSION: The c-kit/PDGFRalpha genotypes after imatinib mesylate resistance may both relate to primary mutations and efficacy of sunitinib treatment. Sunitinib 131-140 platelet derived growth factor receptor alpha Homo sapiens 22-32 22932406-1 2012 OBJECTIVE: To investigate the relationship between secondary mutations of c-kit/PDGFRalpha resistance to imatinib mesylate and the efficacy of sunitinib in patients with gastrointestinal stromal tumor (GIST). Sunitinib 143-152 platelet derived growth factor receptor alpha Homo sapiens 80-90 22344606-0 2012 Sunitinib enhances antitumor effects against chemotherapy-resistant bladder cancer through suppression of ERK1/2 phosphorylation. Sunitinib 0-9 mitogen-activated protein kinase 3 Homo sapiens 118-124 22568998-0 2012 Q-TWiST analysis to estimate overall benefit for patients with metastatic renal cell carcinoma treated in a phase III trial of sunitinib vs interferon-alpha. Sunitinib 127-136 twist family bHLH transcription factor 1 Homo sapiens 0-7 22568998-6 2012 RESULTS: Q-TWiST scores showed that quality-adjusted survival time was greater with sunitinib than with IFN-alpha, even though certain grade 3/4 toxicities occurred more frequently with sunitinib. Sunitinib 84-93 twist family bHLH transcription factor 1 Homo sapiens 11-16 22568998-9 2012 CONCLUSION: Patients randomised to sunitinib had longer clinical benefit, defined as Q-TWiST scores, than patients randomised to IFN-alpha. Sunitinib 35-44 twist family bHLH transcription factor 1 Homo sapiens 85-92 22969964-6 2012 Combination therapy using the multi-tyrosine kinase inhibitors, sorafenib or sunitinib, with S-1 against breast cancer (MX-1 cell line) and NSCLC (NCI-H460 cell line) was significantly superior to either monotherapy (P<0.01). Sunitinib 77-86 proteasome 26S subunit, non-ATPase 1 Homo sapiens 93-96 22314934-0 2012 Antitumor effect of vascular endothelial growth factor inhibitor sunitinib in preclinical models of hepatocellular carcinoma. Sunitinib 65-74 vascular endothelial growth factor A Homo sapiens 20-54 22314934-4 2012 Recently, another VEGF inhibitor, sunitinib, showed survival benefits in HCC hepatitis B-positive patients, but failed to improve survival in HCC hepatitis C-positive patients. Sunitinib 34-43 vascular endothelial growth factor A Homo sapiens 18-22 22314934-9 2012 RESULTS: Sunitinib has the potential to moderately inhibit proliferation in the Huh7.5 cell line, induce p53 in the p53-wild-type cell line SK-hep-1, and to increase the S-phase and the sub-G1 component of the cell cycle in the Hep3B cell line. Sunitinib 9-18 tumor protein p53 Homo sapiens 105-108 22314934-9 2012 RESULTS: Sunitinib has the potential to moderately inhibit proliferation in the Huh7.5 cell line, induce p53 in the p53-wild-type cell line SK-hep-1, and to increase the S-phase and the sub-G1 component of the cell cycle in the Hep3B cell line. Sunitinib 9-18 tumor protein p53 Homo sapiens 116-119 22314934-9 2012 RESULTS: Sunitinib has the potential to moderately inhibit proliferation in the Huh7.5 cell line, induce p53 in the p53-wild-type cell line SK-hep-1, and to increase the S-phase and the sub-G1 component of the cell cycle in the Hep3B cell line. Sunitinib 9-18 DNL-type zinc finger Homo sapiens 143-148 22344606-4 2012 We hypothesized that sunitinib would be effective in bladder cancer as it is an oral inhibitor of multiple receptor tyrosine kinases, including VEGF receptors, platelet derived growth factor (PDGF) receptors and stem cell factor receptor (c-KIT), and is a standard first-line treatment of advanced clear cell renal carcinoma. Sunitinib 21-30 vascular endothelial growth factor A Homo sapiens 168-172 22344606-4 2012 We hypothesized that sunitinib would be effective in bladder cancer as it is an oral inhibitor of multiple receptor tyrosine kinases, including VEGF receptors, platelet derived growth factor (PDGF) receptors and stem cell factor receptor (c-KIT), and is a standard first-line treatment of advanced clear cell renal carcinoma. Sunitinib 21-30 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 275-280 22437942-4 2012 (Function of activation loop tyrosine phosphorylation in the mechanism of c-Kit autoactivation and its implication in sunitinib resistance. Sunitinib 118-127 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 74-79 22344606-5 2012 In the present study, the antiproliferative effects of sunitinib were clearly demonstrated in KK47, KK47/DDP20 and KK47/ADR cell lines in vitro due to the suppression of ERK1/2 phosphorylation. Sunitinib 67-76 mitogen-activated protein kinase 3 Homo sapiens 194-200 22444679-1 2012 Sunitinib (SU11248) is a highly potent tyrosine kinase inhibitor targeting vascular endothelial growth factor receptor (VEGFR). Sunitinib 0-9 kinase insert domain receptor Homo sapiens 76-119 22484816-4 2012 Here, we show that semaphorin 3A (Sema3A) expression overcomes the proinvasive and prometastatic resistance observed upon angiogenesis reduction by the small-molecule tyrosine inhibitor sunitinib in both pancreatic neuroendocrine tumors (PNETs) in RIP-Tag2 mice and cervical carcinomas in HPV16/E2 mice. Sunitinib 186-195 sema domain, immunoglobulin domain (Ig), short basic domain, secreted, (semaphorin) 3A Mus musculus 19-32 22484816-4 2012 Here, we show that semaphorin 3A (Sema3A) expression overcomes the proinvasive and prometastatic resistance observed upon angiogenesis reduction by the small-molecule tyrosine inhibitor sunitinib in both pancreatic neuroendocrine tumors (PNETs) in RIP-Tag2 mice and cervical carcinomas in HPV16/E2 mice. Sunitinib 186-195 sema domain, immunoglobulin domain (Ig), short basic domain, secreted, (semaphorin) 3A Mus musculus 34-40 22484816-5 2012 By improving cancer tissue oxygenation and extending the normalization window, Sema3A counteracted sunitinib-induced activation of HIF-1alpha, Met tyrosine kinase receptor, epithelial-mesenchymal transition (EMT), and other hypoxia-dependent signaling pathways. Sunitinib 99-108 sema domain, immunoglobulin domain (Ig), short basic domain, secreted, (semaphorin) 3A Mus musculus 79-85 22484816-5 2012 By improving cancer tissue oxygenation and extending the normalization window, Sema3A counteracted sunitinib-induced activation of HIF-1alpha, Met tyrosine kinase receptor, epithelial-mesenchymal transition (EMT), and other hypoxia-dependent signaling pathways. Sunitinib 99-108 hypoxia inducible factor 1, alpha subunit Mus musculus 131-141 22048125-3 2012 Injection of sunitinib malate, AMPK inhibitor, to the mice lowered AV formation in their brains. Sunitinib 13-29 protein kinase AMP-activated non-catalytic subunit beta 1 Homo sapiens 31-35 22326847-2 2012 We tested, if the cytotoxic cancer therapy doxorubicin (Doxo) or the anti-angiogenic therapy sunitinib alters viability and VEGF signaling in primary cardiac microvascular endothelial cells (CMEC) and adult rat ventricular myocytes (ARVM). Sunitinib 93-102 vascular endothelial growth factor A Rattus norvegicus 124-128 22326847-14 2012 VEGF receptor 2 amounts were reduced by Doxo and by sunitinib in a dose-dependent manner in both CMEC and ARVM. Sunitinib 52-61 vascular endothelial growth factor A Rattus norvegicus 0-4 22444679-1 2012 Sunitinib (SU11248) is a highly potent tyrosine kinase inhibitor targeting vascular endothelial growth factor receptor (VEGFR). Sunitinib 0-9 kinase insert domain receptor Homo sapiens 121-126 21633954-5 2012 In vitro, sunitinib inhibited PDGFR phosphorylation on MC38-CEA cells at concentrations similar to those biologically available during human treatment. Sunitinib 10-19 CEA cell adhesion molecule 3 Homo sapiens 60-63 22444679-1 2012 Sunitinib (SU11248) is a highly potent tyrosine kinase inhibitor targeting vascular endothelial growth factor receptor (VEGFR). Sunitinib 12-19 kinase insert domain receptor Homo sapiens 76-119 21633954-7 2012 In a model using CEA-Tg mice bearing CEA(+) tumors, continuous sunitinib followed by vaccine increased intratumoral infiltration of antigen-specific T lymphocytes, decreased immunosuppressant T regulatory cells and myeloid-derived suppressor cells, reduced tumor volumes and increased survival. Sunitinib 64-73 carcinoembryonic antigen gene family Mus musculus 17-20 21633954-7 2012 In a model using CEA-Tg mice bearing CEA(+) tumors, continuous sunitinib followed by vaccine increased intratumoral infiltration of antigen-specific T lymphocytes, decreased immunosuppressant T regulatory cells and myeloid-derived suppressor cells, reduced tumor volumes and increased survival. Sunitinib 64-73 carcinoembryonic antigen gene family Mus musculus 37-40 22444679-1 2012 Sunitinib (SU11248) is a highly potent tyrosine kinase inhibitor targeting vascular endothelial growth factor receptor (VEGFR). Sunitinib 12-19 kinase insert domain receptor Homo sapiens 121-126 21882181-5 2012 RESULTS: VEGF SNP -634 genotype was associated with the prevalence and duration of sunitinib-induced hypertension (as defined by systolic pressure >=150 mmHg and/or diastolic pressure >=90 mmHg) in both univariable analysis (P = .03 and .01, respectively) and multivariable analysis, which adjusted for baseline BP and use of antihypertension medication (P = .05 and .02, respectively). Sunitinib 83-92 vascular endothelial growth factor A Homo sapiens 9-13 21882181-9 2012 CONCLUSIONS: In MCCRCC patients treated with sunitinib, VEGF SNP -634 is associated with hypertension and a combination of VEGF SNP 936 and VEGFR2 SNP 889 genotypes is associated with overall survival. Sunitinib 45-54 vascular endothelial growth factor A Homo sapiens 56-60 21898375-2 2012 Everolimus, an oral mTOR inhibitor, and sunitinib, an oral tyrosine kinase inhibitor targeting VEGF, are standard agents in the management of metastatic RCC. Sunitinib 40-49 vascular endothelial growth factor A Homo sapiens 95-99 22179104-1 2012 PURPOSE: Sunitinib is an oral tyrosine kinase inhibitor of VEGF, PDGF, c-KIT, and flt-3 receptors. Sunitinib 9-18 vascular endothelial growth factor A Homo sapiens 59-63 22338018-4 2012 And, recent phase III studies showed both everolimus and sunitinib improved progression-free survival in pancreatic NETs, validating the phosphoinositide 3-kinase/Akt/mTOR pathway and angiogenesis as important targets for further advances. Sunitinib 57-66 AKT serine/threonine kinase 1 Homo sapiens 163-166 22338018-4 2012 And, recent phase III studies showed both everolimus and sunitinib improved progression-free survival in pancreatic NETs, validating the phosphoinositide 3-kinase/Akt/mTOR pathway and angiogenesis as important targets for further advances. Sunitinib 57-66 mechanistic target of rapamycin kinase Homo sapiens 167-171 22238213-0 2012 P-glycoprotein (ABCB1) and breast cancer resistance protein (ABCG2) restrict brain accumulation of the active sunitinib metabolite N-desethyl sunitinib. Sunitinib 110-119 ATP-binding cassette, sub-family B (MDR/TAP), member 1B Mus musculus 16-21 20960028-1 2012 BACKGROUND: Sunitinib is an oral multitargeted tyrosine kinase inhibitor of vascular endothelial growth factor and platelet-derived growth factor receptors, as well as of other receptor types. Sunitinib 12-21 vascular endothelial growth factor A Homo sapiens 76-110 21104107-7 2012 The superior efficacy of sunitinib against GISTs with KIT exon 9 mutations appears to be similar in Korean patients to Western experience although statistical significance was not secured. Sunitinib 25-34 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 54-57 22237457-11 2012 CONCLUSIONS: VEGFR TKIs, especially sunitinib, are active and tolerated by mRCC patients with poor risk features. Sunitinib 36-45 kinase insert domain receptor Homo sapiens 13-18 22238213-0 2012 P-glycoprotein (ABCB1) and breast cancer resistance protein (ABCG2) restrict brain accumulation of the active sunitinib metabolite N-desethyl sunitinib. Sunitinib 110-119 ATP binding cassette subfamily G member 2 (Junior blood group) Mus musculus 61-66 22238213-7 2012 In the absence of the ABCB1 and ABCG2 inhibitor elacridar, brain concentrations of N-desethyl sunitinib were detectable only in Abcb1a/1b(-/-)/Abcg2(-/-) mice after sunitinib administration. Sunitinib 94-103 ATP-binding cassette, sub-family B (MDR/TAP), member 1A Mus musculus 128-134 22238213-7 2012 In the absence of the ABCB1 and ABCG2 inhibitor elacridar, brain concentrations of N-desethyl sunitinib were detectable only in Abcb1a/1b(-/-)/Abcg2(-/-) mice after sunitinib administration. Sunitinib 94-103 ATP binding cassette subfamily G member 2 (Junior blood group) Mus musculus 143-148 21878661-0 2012 Sunitinib induces apoptosis in pheochromocytoma tumor cells by inhibiting VEGFR2/Akt/mTOR/S6K1 pathways through modulation of Bcl-2 and BAD. Sunitinib 0-9 kinase insert domain receptor Rattus norvegicus 74-80 22331954-1 2012 PURPOSE: To investigate whether sunitinib plus docetaxel improves clinical outcomes for patients with human epidermal growth factor receptor 2 (HER2)/neu-negative advanced breast cancer (ABC) versus docetaxel alone. Sunitinib 32-41 erb-b2 receptor tyrosine kinase 2 Homo sapiens 108-142 22331954-1 2012 PURPOSE: To investigate whether sunitinib plus docetaxel improves clinical outcomes for patients with human epidermal growth factor receptor 2 (HER2)/neu-negative advanced breast cancer (ABC) versus docetaxel alone. Sunitinib 32-41 erb-b2 receptor tyrosine kinase 2 Homo sapiens 144-148 21878661-0 2012 Sunitinib induces apoptosis in pheochromocytoma tumor cells by inhibiting VEGFR2/Akt/mTOR/S6K1 pathways through modulation of Bcl-2 and BAD. Sunitinib 0-9 AKT serine/threonine kinase 1 Rattus norvegicus 81-84 21878661-0 2012 Sunitinib induces apoptosis in pheochromocytoma tumor cells by inhibiting VEGFR2/Akt/mTOR/S6K1 pathways through modulation of Bcl-2 and BAD. Sunitinib 0-9 mechanistic target of rapamycin kinase Rattus norvegicus 85-89 21878661-0 2012 Sunitinib induces apoptosis in pheochromocytoma tumor cells by inhibiting VEGFR2/Akt/mTOR/S6K1 pathways through modulation of Bcl-2 and BAD. Sunitinib 0-9 ribosomal protein S6 kinase B1 Rattus norvegicus 90-94 21878661-0 2012 Sunitinib induces apoptosis in pheochromocytoma tumor cells by inhibiting VEGFR2/Akt/mTOR/S6K1 pathways through modulation of Bcl-2 and BAD. Sunitinib 0-9 BCL2, apoptosis regulator Rattus norvegicus 126-131 21878661-1 2012 Sunitinib is an oral multitargeted receptor tyrosine kinase inhibitor with antiangiogenic and antitumor activity that mainly targets vascular endothelial growth factor receptors (VEGFRs). Sunitinib 0-9 kinase insert domain receptor Rattus norvegicus 179-185 21878661-5 2012 Furthermore, in support of these findings, we found that sunitinib induced a reduction in the expression of the antiapoptotic molecule Bcl-2 as well as dephosphorylation of the proapoptotic molecule BAD, which results in the activation of BAD in these cells. Sunitinib 57-66 BCL2, apoptosis regulator Rattus norvegicus 135-140 21878661-6 2012 Consistent with these apoptotic effects, our results showed that sunitinib inhibited phosphorylation of Akt and mTOR and was followed by a reduction of S6K1, which is a well-known target of mTOR. Sunitinib 65-74 AKT serine/threonine kinase 1 Rattus norvegicus 104-107 21878661-6 2012 Consistent with these apoptotic effects, our results showed that sunitinib inhibited phosphorylation of Akt and mTOR and was followed by a reduction of S6K1, which is a well-known target of mTOR. Sunitinib 65-74 mechanistic target of rapamycin kinase Rattus norvegicus 112-116 21878661-6 2012 Consistent with these apoptotic effects, our results showed that sunitinib inhibited phosphorylation of Akt and mTOR and was followed by a reduction of S6K1, which is a well-known target of mTOR. Sunitinib 65-74 ribosomal protein S6 kinase B1 Rattus norvegicus 152-156 21878661-6 2012 Consistent with these apoptotic effects, our results showed that sunitinib inhibited phosphorylation of Akt and mTOR and was followed by a reduction of S6K1, which is a well-known target of mTOR. Sunitinib 65-74 mechanistic target of rapamycin kinase Rattus norvegicus 190-194 21878661-7 2012 Knockdown of VEGFR-2 attenuated the sunitinib-induced effects, including apoptosis and inhibition of signaling pathways such as the phosphorylation of Akt as well as mTOR, and Bcl-2, which confirmed that these effects could be mediated by VEGFR-2. Sunitinib 36-45 kinase insert domain receptor Rattus norvegicus 13-20 21878661-7 2012 Knockdown of VEGFR-2 attenuated the sunitinib-induced effects, including apoptosis and inhibition of signaling pathways such as the phosphorylation of Akt as well as mTOR, and Bcl-2, which confirmed that these effects could be mediated by VEGFR-2. Sunitinib 36-45 AKT serine/threonine kinase 1 Rattus norvegicus 151-154 21878661-7 2012 Knockdown of VEGFR-2 attenuated the sunitinib-induced effects, including apoptosis and inhibition of signaling pathways such as the phosphorylation of Akt as well as mTOR, and Bcl-2, which confirmed that these effects could be mediated by VEGFR-2. Sunitinib 36-45 mechanistic target of rapamycin kinase Rattus norvegicus 166-170 21878661-7 2012 Knockdown of VEGFR-2 attenuated the sunitinib-induced effects, including apoptosis and inhibition of signaling pathways such as the phosphorylation of Akt as well as mTOR, and Bcl-2, which confirmed that these effects could be mediated by VEGFR-2. Sunitinib 36-45 BCL2, apoptosis regulator Rattus norvegicus 176-181 21878661-7 2012 Knockdown of VEGFR-2 attenuated the sunitinib-induced effects, including apoptosis and inhibition of signaling pathways such as the phosphorylation of Akt as well as mTOR, and Bcl-2, which confirmed that these effects could be mediated by VEGFR-2. Sunitinib 36-45 kinase insert domain receptor Rattus norvegicus 239-246 21878661-8 2012 In addition, silencing of S6K1 induced apoptosis accompanied by a decrease in the phosphorylation of BAD and Bcl-2, similar to that observed with sunitinib treatment. Sunitinib 146-155 ribosomal protein S6 kinase B1 Rattus norvegicus 26-30 21878661-9 2012 Thus, these results together suggest that sunitinib initially exerts its apoptotic effect through the inhibition of VEGFR-2, which, when followed by reduction of its downstream effectors, including Akt/mTOR/S6K1, may lead to inhibition of the antiapoptotic molecule Bcl-2 and activation of the proapoptotic molecule BAD in PC12 cells. Sunitinib 42-51 kinase insert domain receptor Rattus norvegicus 116-123 21878661-9 2012 Thus, these results together suggest that sunitinib initially exerts its apoptotic effect through the inhibition of VEGFR-2, which, when followed by reduction of its downstream effectors, including Akt/mTOR/S6K1, may lead to inhibition of the antiapoptotic molecule Bcl-2 and activation of the proapoptotic molecule BAD in PC12 cells. Sunitinib 42-51 AKT serine/threonine kinase 1 Rattus norvegicus 198-201 21878661-9 2012 Thus, these results together suggest that sunitinib initially exerts its apoptotic effect through the inhibition of VEGFR-2, which, when followed by reduction of its downstream effectors, including Akt/mTOR/S6K1, may lead to inhibition of the antiapoptotic molecule Bcl-2 and activation of the proapoptotic molecule BAD in PC12 cells. Sunitinib 42-51 mechanistic target of rapamycin kinase Rattus norvegicus 202-206 21878661-9 2012 Thus, these results together suggest that sunitinib initially exerts its apoptotic effect through the inhibition of VEGFR-2, which, when followed by reduction of its downstream effectors, including Akt/mTOR/S6K1, may lead to inhibition of the antiapoptotic molecule Bcl-2 and activation of the proapoptotic molecule BAD in PC12 cells. Sunitinib 42-51 ribosomal protein S6 kinase B1 Rattus norvegicus 207-211 21878661-9 2012 Thus, these results together suggest that sunitinib initially exerts its apoptotic effect through the inhibition of VEGFR-2, which, when followed by reduction of its downstream effectors, including Akt/mTOR/S6K1, may lead to inhibition of the antiapoptotic molecule Bcl-2 and activation of the proapoptotic molecule BAD in PC12 cells. Sunitinib 42-51 BCL2, apoptosis regulator Rattus norvegicus 266-271 22585997-5 2012 A similar benefit was found in orthotopic Panc-1 pancreatic carcinomas treated with sunitinib plus PF-04217903 and in RIP-Tag2 tumors treated with XL184 (cabozantinib), which simultaneously blocks VEGF and c-Met signaling. Sunitinib 84-93 pancreas protein 1 Mus musculus 42-48 22369443-8 2012 However, the decision to use either high-dose imatinib or sunitinib should be based on the underlying cause of failure on imatinib, KIT mutational status and on whether the patient is intolerant of or has developed a resistance to imatinib. Sunitinib 58-67 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 132-135 22399609-4 2012 RESULTS: Mono-Mac 1 cells had inhibited proliferation and extracellular-signal regulated kinase activity as a result of CSF-1R inhibition and a dose-dependent increase in CSF-1R expression with both sunitinib and cFMS-I. Sunitinib 199-208 integrin subunit alpha M Homo sapiens 14-19 22399609-4 2012 RESULTS: Mono-Mac 1 cells had inhibited proliferation and extracellular-signal regulated kinase activity as a result of CSF-1R inhibition and a dose-dependent increase in CSF-1R expression with both sunitinib and cFMS-I. Sunitinib 199-208 colony stimulating factor 1 receptor Homo sapiens 171-177 22399609-5 2012 CONCLUSION: Our results suggest potential for CSF-1R as an important target of sunitinib or other similar drugs. Sunitinib 79-88 colony stimulating factor 1 receptor Homo sapiens 46-52 22261812-0 2012 Sunitinib therapy for melanoma patients with KIT mutations. Sunitinib 0-9 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 45-48 22261812-2 2012 We aimed to assess the predictive role of KIT mutation, amplification, or overexpression for response to treatment with the kinase inhibitor sunitinib. Sunitinib 141-150 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 42-45 22261812-4 2012 Patients with mutations, amplifications, or overexpression of KIT were treated with sunitinib and responses measured by Response Evaluation Criteria in Solid Tumors (RECIST). Sunitinib 84-93 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 62-65 22261812-11 2012 CONCLUSIONS: Sunitinib may have activity in patients with melanoma and KIT mutations; more study is needed. Sunitinib 13-22 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 71-74 22369324-9 2012 presented retrospective data showing that retreatment with VEGF-directed targeted agents, including sunitinib, bevacizumab/interferon, dovitinib and sorafenib, was associated with a progression-free survival time of approximately 5 months. Sunitinib 100-109 vascular endothelial growth factor A Homo sapiens 59-63 21445973-5 2012 Examination of intracellular signaling found that Akt/ mammalian target of rapamycin (mTOR) signaling remained activated in GIST-T1R but not in parental GIST-T1 cells, after exposure of these cells to sunitinib, as measured by immunoblotting. Sunitinib 201-210 mechanistic target of rapamycin kinase Homo sapiens 55-84 22115851-0 2012 RhoB mediates antitumor synergy of combined ixabepilone and sunitinib in human ovarian serous cancer. Sunitinib 60-69 ras homolog family member B Homo sapiens 0-4 22115851-6 2012 The GTPase, RhoB, was synergistically upregulated in cells treated with ixabepilone and sunitinib. Sunitinib 88-97 ras homolog family member B Homo sapiens 12-16 22115851-9 2012 CONCLUSIONS: Ixabepilone plus sunitinib demonstrated antitumor synergy via RhoB in naive and paclitaxel-resistant cells resulting in apoptosis. Sunitinib 30-39 ras homolog family member B Homo sapiens 75-79 22115851-11 2012 RhoB could be envisioned as an early biomarker of response to therapy in a planned Phase II clinical trial to assess the efficacy of ixabepilone combined with a receptor tyrosine kinase inhibitor such as sunitinib. Sunitinib 204-213 ras homolog family member B Homo sapiens 0-4 25806151-3 2012 Thus, this system has become the focus of therapeutic interventions, which led to the approval of the anti-VEGF blocking antibody bevacizumab and the VEGFR-2 pathway inhibitors pazopanib, sorafenib and sunitinib. Sunitinib 202-211 vascular endothelial growth factor A Homo sapiens 107-111 25806151-3 2012 Thus, this system has become the focus of therapeutic interventions, which led to the approval of the anti-VEGF blocking antibody bevacizumab and the VEGFR-2 pathway inhibitors pazopanib, sorafenib and sunitinib. Sunitinib 202-211 kinase insert domain receptor Homo sapiens 150-157 22308314-6 2012 We further show that the Akt/beta-catenin cancer stem cell regulatory pathway is activated in breast cancer cells under hypoxic conditions in vitro and in sunitinib-treated mouse xenografts. Sunitinib 155-164 thymoma viral proto-oncogene 1 Mus musculus 25-28 22308314-6 2012 We further show that the Akt/beta-catenin cancer stem cell regulatory pathway is activated in breast cancer cells under hypoxic conditions in vitro and in sunitinib-treated mouse xenografts. Sunitinib 155-164 catenin (cadherin associated protein), beta 1 Mus musculus 29-41 21445973-0 2012 Long-term exposure of gastrointestinal stromal tumor cells to sunitinib induces epigenetic silencing of the PTEN gene. Sunitinib 62-71 phosphatase and tensin homolog Homo sapiens 108-112 21445973-5 2012 Examination of intracellular signaling found that Akt/ mammalian target of rapamycin (mTOR) signaling remained activated in GIST-T1R but not in parental GIST-T1 cells, after exposure of these cells to sunitinib, as measured by immunoblotting. Sunitinib 201-210 AKT serine/threonine kinase 1 Homo sapiens 50-53 21445973-5 2012 Examination of intracellular signaling found that Akt/ mammalian target of rapamycin (mTOR) signaling remained activated in GIST-T1R but not in parental GIST-T1 cells, after exposure of these cells to sunitinib, as measured by immunoblotting. Sunitinib 201-210 mechanistic target of rapamycin kinase Homo sapiens 86-90 21445973-7 2012 Notably, forced-expression of PTEN in GIST-T1R cells negatively regulated the Akt/mTOR pathways and sensitized these cells to sunitinib-mediated growth arrest and apoptosis. Sunitinib 126-135 phosphatase and tensin homolog Homo sapiens 30-34 21445974-0 2012 Inhibition of activated receptor tyrosine kinases by Sunitinib induces growth arrest and sensitizes melanoma cells to Bortezomib by blocking Akt pathway. Sunitinib 53-62 AKT serine/threonine kinase 1 Homo sapiens 141-144 21445974-6 2012 Moreover, the two metastatic melanoma cell lines harbored an activated receptor (PDGFRalpha and VEGFR, respectively), and Sunitinib suppressed the phosphorylation of the RTKs and their downstream targets Akt and ribosomal protein S6, in these two cell lines. Sunitinib 122-131 kinase insert domain receptor Homo sapiens 96-101 21445974-6 2012 Moreover, the two metastatic melanoma cell lines harbored an activated receptor (PDGFRalpha and VEGFR, respectively), and Sunitinib suppressed the phosphorylation of the RTKs and their downstream targets Akt and ribosomal protein S6, in these two cell lines. Sunitinib 122-131 AKT serine/threonine kinase 1 Homo sapiens 204-207 21445974-6 2012 Moreover, the two metastatic melanoma cell lines harbored an activated receptor (PDGFRalpha and VEGFR, respectively), and Sunitinib suppressed the phosphorylation of the RTKs and their downstream targets Akt and ribosomal protein S6, in these two cell lines. Sunitinib 122-131 ribosomal protein S6 Homo sapiens 212-232 21445974-10 2012 Moreover, LY294002, a PI3K inhibitor, sensitized melanoma cells to Bortezomib treatment, suggesting that downregulation of phospho-Akt by Sunitinib mediates the synergy obtained by Bortezomib + Sunitinib cotreatment. Sunitinib 138-147 AKT serine/threonine kinase 1 Homo sapiens 131-134 21445974-10 2012 Moreover, LY294002, a PI3K inhibitor, sensitized melanoma cells to Bortezomib treatment, suggesting that downregulation of phospho-Akt by Sunitinib mediates the synergy obtained by Bortezomib + Sunitinib cotreatment. Sunitinib 194-203 AKT serine/threonine kinase 1 Homo sapiens 131-134 22395469-8 2012 After PTK/ZK or sunitinib treatment, no-VEGFR-methylation cancer cells showed dose- or time-dependent decreases in proliferation. Sunitinib 16-25 kinase insert domain receptor Homo sapiens 40-45 22866542-0 2012 Structural and functional analysis of KIT gene encoding receptor tyrosine kinase and its interaction with sunitinib and HDAC inhibitors: an in silico approach. Sunitinib 106-115 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 38-41 22078005-2 2012 Since then, vascular endothelial growth factor (VEGF) has been identified as the most potent cytokine to induce angiogenesis and drugs targeting VEGF, principally the humanized monoclonal antibody bevacizumab and the tyrosine kinase inhibitors sunitinib and sorafenib, have proven therapeutic benefit. Sunitinib 244-253 vascular endothelial growth factor A Homo sapiens 12-46 22078005-2 2012 Since then, vascular endothelial growth factor (VEGF) has been identified as the most potent cytokine to induce angiogenesis and drugs targeting VEGF, principally the humanized monoclonal antibody bevacizumab and the tyrosine kinase inhibitors sunitinib and sorafenib, have proven therapeutic benefit. Sunitinib 244-253 vascular endothelial growth factor A Homo sapiens 48-52 22450834-6 2012 After one course of sunitinib, mental status was altered due to hypercalcemia (serum calcium level was 18.6 mg/dl and PTH-rP was 3.7 pmol/l). Sunitinib 20-29 parathyroid hormone like hormone Homo sapiens 118-124 22866542-0 2012 Structural and functional analysis of KIT gene encoding receptor tyrosine kinase and its interaction with sunitinib and HDAC inhibitors: an in silico approach. Sunitinib 106-115 ret proto-oncogene Homo sapiens 56-80 22866542-4 2012 In this study, a computational analysis to detect the most deleterious nonsynonymous SNPs of KIT gene was performed and investigated its binding affinity to native and predicted mutant protein structure (D816V) with sunitinib and HDAC (Trichostatin A and Panobinostat) inhibitors was investigated. Sunitinib 216-225 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 93-96 24451769-4 2012 Further, vascular endothelial growth factor receptor (VEGF-R) inhibitory agents including sorafenib, sunitinib, pazopanib, and axitinib that are already approved in the United States for use in advanced renal cell carcinoma have shown high response rates in treating advanced DTCs in multiple phase II trials, and have become commonly used in progressive radioiodine-refractory metastatic DTC. Sunitinib 101-110 kinase insert domain receptor Homo sapiens 9-52 22497103-15 2012 Sunitinib, recommended by NICE, is administered orally and acts by inhibiting the VEGF receptor. Sunitinib 0-9 vascular endothelial growth factor A Homo sapiens 82-86 22191389-2 2012 The multikinase inhibitor Sunitinib has been shown to inhibit the kinase activity of the RET oncogene and reduce proliferation in differentiated thyroid cancer cells harboring the RET/PTC rearrangement. Sunitinib 26-35 ret proto-oncogene Homo sapiens 89-92 22191389-2 2012 The multikinase inhibitor Sunitinib has been shown to inhibit the kinase activity of the RET oncogene and reduce proliferation in differentiated thyroid cancer cells harboring the RET/PTC rearrangement. Sunitinib 26-35 ret proto-oncogene Homo sapiens 180-183 22191389-5 2012 RESULTS: Exposure to nanomolar concentrations of Sunitinib significantly reduced cell viability in only TPC-1 cells, and this effect was paralleled by reduction of cyclin D1 levels. Sunitinib 49-58 two pore segment channel 1 Homo sapiens 104-109 22191389-5 2012 RESULTS: Exposure to nanomolar concentrations of Sunitinib significantly reduced cell viability in only TPC-1 cells, and this effect was paralleled by reduction of cyclin D1 levels. Sunitinib 49-58 cyclin D1 Homo sapiens 164-173 21933194-7 2012 Spleen cells from sunitinib-treated mice contained smaller numbers of antigen-specific IgG-producing cells and secreted lower amounts of both Th1 and Th2 cytokines than those of the control mice, whereas the levels of antigen-specific antibodies in serum were not decreased. Sunitinib 18-27 negative elongation factor complex member C/D, Th1l Mus musculus 142-145 21933194-7 2012 Spleen cells from sunitinib-treated mice contained smaller numbers of antigen-specific IgG-producing cells and secreted lower amounts of both Th1 and Th2 cytokines than those of the control mice, whereas the levels of antigen-specific antibodies in serum were not decreased. Sunitinib 18-27 heart and neural crest derivatives expressed 2 Mus musculus 150-153 21933194-8 2012 The reactions and MMCP-1 release in oral antigen-induced anaphylaxis and passive systemic anaphylaxis were attenuated even by a single predose of sunitinib. Sunitinib 146-155 mast cell protease 9 Mus musculus 18-24 24451769-4 2012 Further, vascular endothelial growth factor receptor (VEGF-R) inhibitory agents including sorafenib, sunitinib, pazopanib, and axitinib that are already approved in the United States for use in advanced renal cell carcinoma have shown high response rates in treating advanced DTCs in multiple phase II trials, and have become commonly used in progressive radioiodine-refractory metastatic DTC. Sunitinib 101-110 kinase insert domain receptor Homo sapiens 54-60 21699503-2 2012 Indolinones (e.g. SU5416 and Sutent) and anilinophthalazines (e.g. PTK787) are potent small molecule inhibitors of VEGFR2 and other tyrosine kinases, but their effects on VEGF-A- and bFGF-stimulated endothelial responses are unclear. Sunitinib 29-35 kinase insert domain receptor Homo sapiens 115-121 21699503-2 2012 Indolinones (e.g. SU5416 and Sutent) and anilinophthalazines (e.g. PTK787) are potent small molecule inhibitors of VEGFR2 and other tyrosine kinases, but their effects on VEGF-A- and bFGF-stimulated endothelial responses are unclear. Sunitinib 29-35 vascular endothelial growth factor A Homo sapiens 171-177 21699503-2 2012 Indolinones (e.g. SU5416 and Sutent) and anilinophthalazines (e.g. PTK787) are potent small molecule inhibitors of VEGFR2 and other tyrosine kinases, but their effects on VEGF-A- and bFGF-stimulated endothelial responses are unclear. Sunitinib 29-35 fibroblast growth factor 2 Homo sapiens 183-187 21699503-7 2012 Sutent is a potent inhibitor of both VEGFR2 and FGFR1 tyrosine kinase activity in vitro. Sunitinib 0-6 kinase insert domain receptor Homo sapiens 37-43 21699503-7 2012 Sutent is a potent inhibitor of both VEGFR2 and FGFR1 tyrosine kinase activity in vitro. Sunitinib 0-6 fibroblast growth factor receptor 1 Homo sapiens 48-53 22673043-0 2012 Impact of genetic variation in breast cancer resistance protein (BCRP/ABCG2) on sunitinib pharmacokinetics. Sunitinib 80-89 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 31-63 22814298-6 2012 A 48 h exposure to sunitinib (10 microM) significantly decreased forward scatter and increased annexin-V-binding, effects paralleled by significant increase of [Ca(2+)](i). Sunitinib 19-28 annexin A5 Homo sapiens 95-104 22814298-8 2012 Sunitinib induced annexin-V-binding was slightly, but significantly blunted by removal of extracellular Ca(2+), by p38 kinase inhibitor SB203580 (10 microM) and by the pancaspase inhibitor zVAD (10 microM). Sunitinib 0-9 annexin A5 Homo sapiens 18-27 22814298-8 2012 Sunitinib induced annexin-V-binding was slightly, but significantly blunted by removal of extracellular Ca(2+), by p38 kinase inhibitor SB203580 (10 microM) and by the pancaspase inhibitor zVAD (10 microM). Sunitinib 0-9 mitogen-activated protein kinase 14 Homo sapiens 115-118 22015552-10 2012 More recently, sunitinib (Sutent, Pfizer, New York, NY), which inhibits vascular endothelial growth factor receptor (VEGFR) in addition to KIT and PDGFRA, has proven efficacious in patients who are intolerant or refractory to imatinib. Sunitinib 15-24 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 139-142 22015552-10 2012 More recently, sunitinib (Sutent, Pfizer, New York, NY), which inhibits vascular endothelial growth factor receptor (VEGFR) in addition to KIT and PDGFRA, has proven efficacious in patients who are intolerant or refractory to imatinib. Sunitinib 15-24 platelet derived growth factor receptor alpha Homo sapiens 147-153 22015552-10 2012 More recently, sunitinib (Sutent, Pfizer, New York, NY), which inhibits vascular endothelial growth factor receptor (VEGFR) in addition to KIT and PDGFRA, has proven efficacious in patients who are intolerant or refractory to imatinib. Sunitinib 26-32 kinase insert domain receptor Homo sapiens 72-115 22015552-10 2012 More recently, sunitinib (Sutent, Pfizer, New York, NY), which inhibits vascular endothelial growth factor receptor (VEGFR) in addition to KIT and PDGFRA, has proven efficacious in patients who are intolerant or refractory to imatinib. Sunitinib 26-32 kinase insert domain receptor Homo sapiens 117-122 22015552-10 2012 More recently, sunitinib (Sutent, Pfizer, New York, NY), which inhibits vascular endothelial growth factor receptor (VEGFR) in addition to KIT and PDGFRA, has proven efficacious in patients who are intolerant or refractory to imatinib. Sunitinib 26-32 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 139-142 22015552-10 2012 More recently, sunitinib (Sutent, Pfizer, New York, NY), which inhibits vascular endothelial growth factor receptor (VEGFR) in addition to KIT and PDGFRA, has proven efficacious in patients who are intolerant or refractory to imatinib. Sunitinib 26-32 platelet derived growth factor receptor alpha Homo sapiens 147-153 22855636-2 2012 Early events in GIST development are activating mutations in KIT or PDGFRA, which occur in most GISTs and encode for mutated tyrosine receptor kinases that are therapeutic targets for tyrosine kinase inhibitors, including imatinib and sunitinib. Sunitinib 235-244 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 61-64 22855636-2 2012 Early events in GIST development are activating mutations in KIT or PDGFRA, which occur in most GISTs and encode for mutated tyrosine receptor kinases that are therapeutic targets for tyrosine kinase inhibitors, including imatinib and sunitinib. Sunitinib 235-244 platelet derived growth factor receptor alpha Homo sapiens 68-74 22015552-10 2012 More recently, sunitinib (Sutent, Pfizer, New York, NY), which inhibits vascular endothelial growth factor receptor (VEGFR) in addition to KIT and PDGFRA, has proven efficacious in patients who are intolerant or refractory to imatinib. Sunitinib 15-24 kinase insert domain receptor Homo sapiens 72-115 22015552-10 2012 More recently, sunitinib (Sutent, Pfizer, New York, NY), which inhibits vascular endothelial growth factor receptor (VEGFR) in addition to KIT and PDGFRA, has proven efficacious in patients who are intolerant or refractory to imatinib. Sunitinib 15-24 kinase insert domain receptor Homo sapiens 117-122 22673043-0 2012 Impact of genetic variation in breast cancer resistance protein (BCRP/ABCG2) on sunitinib pharmacokinetics. Sunitinib 80-89 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 65-69 22673043-0 2012 Impact of genetic variation in breast cancer resistance protein (BCRP/ABCG2) on sunitinib pharmacokinetics. Sunitinib 80-89 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 70-75 22673043-1 2012 To elucidate the impact of genetic variations in breast cancer resistance protein (BCRP/ABCG2) and P-glycoprotein (MDR1/ABCB1) on the pharmacokinetics of sunitinib, we carried out a pharmacogenetic study in a clinical setting and pharmacokinetic analysis using Abcg2(-/-), Abcb1a/1b(-/-) and Abcb1a/1b;Abcg2(-/-) mice. Sunitinib 154-163 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 49-81 22673043-1 2012 To elucidate the impact of genetic variations in breast cancer resistance protein (BCRP/ABCG2) and P-glycoprotein (MDR1/ABCB1) on the pharmacokinetics of sunitinib, we carried out a pharmacogenetic study in a clinical setting and pharmacokinetic analysis using Abcg2(-/-), Abcb1a/1b(-/-) and Abcb1a/1b;Abcg2(-/-) mice. Sunitinib 154-163 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 83-87 22673043-1 2012 To elucidate the impact of genetic variations in breast cancer resistance protein (BCRP/ABCG2) and P-glycoprotein (MDR1/ABCB1) on the pharmacokinetics of sunitinib, we carried out a pharmacogenetic study in a clinical setting and pharmacokinetic analysis using Abcg2(-/-), Abcb1a/1b(-/-) and Abcb1a/1b;Abcg2(-/-) mice. Sunitinib 154-163 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 88-93 22673043-1 2012 To elucidate the impact of genetic variations in breast cancer resistance protein (BCRP/ABCG2) and P-glycoprotein (MDR1/ABCB1) on the pharmacokinetics of sunitinib, we carried out a pharmacogenetic study in a clinical setting and pharmacokinetic analysis using Abcg2(-/-), Abcb1a/1b(-/-) and Abcb1a/1b;Abcg2(-/-) mice. Sunitinib 154-163 ATP binding cassette subfamily B member 1 Homo sapiens 99-113 22673043-1 2012 To elucidate the impact of genetic variations in breast cancer resistance protein (BCRP/ABCG2) and P-glycoprotein (MDR1/ABCB1) on the pharmacokinetics of sunitinib, we carried out a pharmacogenetic study in a clinical setting and pharmacokinetic analysis using Abcg2(-/-), Abcb1a/1b(-/-) and Abcb1a/1b;Abcg2(-/-) mice. Sunitinib 154-163 ATP binding cassette subfamily B member 1 Homo sapiens 115-119 22673043-1 2012 To elucidate the impact of genetic variations in breast cancer resistance protein (BCRP/ABCG2) and P-glycoprotein (MDR1/ABCB1) on the pharmacokinetics of sunitinib, we carried out a pharmacogenetic study in a clinical setting and pharmacokinetic analysis using Abcg2(-/-), Abcb1a/1b(-/-) and Abcb1a/1b;Abcg2(-/-) mice. Sunitinib 154-163 ATP binding cassette subfamily B member 1 Homo sapiens 120-125 22673043-1 2012 To elucidate the impact of genetic variations in breast cancer resistance protein (BCRP/ABCG2) and P-glycoprotein (MDR1/ABCB1) on the pharmacokinetics of sunitinib, we carried out a pharmacogenetic study in a clinical setting and pharmacokinetic analysis using Abcg2(-/-), Abcb1a/1b(-/-) and Abcb1a/1b;Abcg2(-/-) mice. Sunitinib 154-163 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 261-266 22673043-1 2012 To elucidate the impact of genetic variations in breast cancer resistance protein (BCRP/ABCG2) and P-glycoprotein (MDR1/ABCB1) on the pharmacokinetics of sunitinib, we carried out a pharmacogenetic study in a clinical setting and pharmacokinetic analysis using Abcg2(-/-), Abcb1a/1b(-/-) and Abcb1a/1b;Abcg2(-/-) mice. Sunitinib 154-163 ATP-binding cassette, sub-family B (MDR/TAP), member 1A Mus musculus 273-279 22673043-1 2012 To elucidate the impact of genetic variations in breast cancer resistance protein (BCRP/ABCG2) and P-glycoprotein (MDR1/ABCB1) on the pharmacokinetics of sunitinib, we carried out a pharmacogenetic study in a clinical setting and pharmacokinetic analysis using Abcg2(-/-), Abcb1a/1b(-/-) and Abcb1a/1b;Abcg2(-/-) mice. Sunitinib 154-163 ATP-binding cassette, sub-family B (MDR/TAP), member 1A Mus musculus 292-298 22673043-1 2012 To elucidate the impact of genetic variations in breast cancer resistance protein (BCRP/ABCG2) and P-glycoprotein (MDR1/ABCB1) on the pharmacokinetics of sunitinib, we carried out a pharmacogenetic study in a clinical setting and pharmacokinetic analysis using Abcg2(-/-), Abcb1a/1b(-/-) and Abcb1a/1b;Abcg2(-/-) mice. Sunitinib 154-163 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 302-307 22673043-5 2012 The dose-adjusted AUC(0-24) of sunitinib was significantly higher in patients with a heterozygous variant for ABCG2 421C>A than in wild-type patients (p = 0.02), and one homozygous patient showed the highest dose-adjusted AUC(0-24). Sunitinib 31-40 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 110-115 22673043-7 2012 The maximum concentration and AUC(0-4) of sunitinib were significantly higher in Abcg2(-/-), Abcb1a/1b(-/-) and Abcb1a/1b;Abcg2(-/-) mice than wild-type mice when sunitinib was given orally but not intraperitoneally. Sunitinib 42-51 ATP binding cassette subfamily G member 2 (Junior blood group) Mus musculus 81-86 22673043-7 2012 The maximum concentration and AUC(0-4) of sunitinib were significantly higher in Abcg2(-/-), Abcb1a/1b(-/-) and Abcb1a/1b;Abcg2(-/-) mice than wild-type mice when sunitinib was given orally but not intraperitoneally. Sunitinib 42-51 ATP-binding cassette, sub-family B (MDR/TAP), member 1A Mus musculus 93-99 22673043-7 2012 The maximum concentration and AUC(0-4) of sunitinib were significantly higher in Abcg2(-/-), Abcb1a/1b(-/-) and Abcb1a/1b;Abcg2(-/-) mice than wild-type mice when sunitinib was given orally but not intraperitoneally. Sunitinib 42-51 ATP-binding cassette, sub-family B (MDR/TAP), member 1A Mus musculus 112-118 22673043-7 2012 The maximum concentration and AUC(0-4) of sunitinib were significantly higher in Abcg2(-/-), Abcb1a/1b(-/-) and Abcb1a/1b;Abcg2(-/-) mice than wild-type mice when sunitinib was given orally but not intraperitoneally. Sunitinib 42-51 ATP binding cassette subfamily G member 2 (Junior blood group) Mus musculus 122-127 22673043-9 2012 These results suggest that the loss of protein expression of ABCG2 by genetic polymorphism is associated with an increase in the systemic exposure to sunitinib and sunitinib-induced toxicity. Sunitinib 150-159 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 61-66 22673043-9 2012 These results suggest that the loss of protein expression of ABCG2 by genetic polymorphism is associated with an increase in the systemic exposure to sunitinib and sunitinib-induced toxicity. Sunitinib 164-173 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 61-66 21898502-10 2012 In addition, sunitinib treatment of tumor-bearing mice was associated with suppression of STAT3 and a block in T-cell tolerance. Sunitinib 13-22 signal transducer and activator of transcription 3 Mus musculus 90-95 21898502-11 2012 CONCLUSION: These findings indicate that sunitinib inhibits HCC tumor growth directly through the STAT3 pathway and prevents tumor antigen-specific CD8(+) T-cell tolerance, thus defining a synergistic chemoimmunotherapeutic approach for HCC. Sunitinib 41-50 signal transducer and activator of transcription 3 Mus musculus 98-103 22677881-8 2012 Predominantly the superiority of sunitinib regarding PFS2 contributed to the longer combined PFS in sequential use. Sunitinib 33-42 GINS complex subunit 2 Homo sapiens 53-57 23017141-1 2012 The receptor tyrosine kinase inhibitor, sunitinib, is astonishingly effective in its capacity to reduce MDSCs in peripheral tissues such as blood (human) and spleen (mouse), restoring responsiveness of bystander T lymphocytes to TcR stimulation. Sunitinib 40-49 T cell receptor alpha variable 6-3 Mus musculus 229-232 23017141-5 2012 The presence or absence of GM-CSF is likely the major determinant in each compartment, given that GM-CSF"s capacity to preempt STAT3-dependent with dominant STAT5-dependent hematopoietic programming confers sunitinib resistance and redirects differentiation from the n-MDSC lineage to the more versatile monocytoid (m-MDSC) lineage. Sunitinib 207-216 colony stimulating factor 2 Homo sapiens 98-104 21351087-0 2012 Brain accumulation of sunitinib is restricted by P-glycoprotein (ABCB1) and breast cancer resistance protein (ABCG2) and can be enhanced by oral elacridar and sunitinib coadministration. Sunitinib 22-31 ATP-binding cassette, sub-family B (MDR/TAP), member 1B Mus musculus 65-70 21351087-0 2012 Brain accumulation of sunitinib is restricted by P-glycoprotein (ABCB1) and breast cancer resistance protein (ABCG2) and can be enhanced by oral elacridar and sunitinib coadministration. Sunitinib 22-31 ATP binding cassette subfamily G member 2 (Junior blood group) Mus musculus 110-115 21351087-2 2012 We aimed to investigate the in vivo roles of the ATP-binding cassette drug efflux transporters ABCB1 and ABCG2 in plasma pharmacokinetics and brain accumulation of oral sunitinib, and the feasibility of improving sunitinib kinetics using oral coadministration of the dual ABCB1/ABCG2 inhibitor elacridar. Sunitinib 169-178 ATP-binding cassette, sub-family B (MDR/TAP), member 1B Mus musculus 95-100 21351087-2 2012 We aimed to investigate the in vivo roles of the ATP-binding cassette drug efflux transporters ABCB1 and ABCG2 in plasma pharmacokinetics and brain accumulation of oral sunitinib, and the feasibility of improving sunitinib kinetics using oral coadministration of the dual ABCB1/ABCG2 inhibitor elacridar. Sunitinib 169-178 ATP binding cassette subfamily G member 2 (Junior blood group) Mus musculus 105-110 21351087-2 2012 We aimed to investigate the in vivo roles of the ATP-binding cassette drug efflux transporters ABCB1 and ABCG2 in plasma pharmacokinetics and brain accumulation of oral sunitinib, and the feasibility of improving sunitinib kinetics using oral coadministration of the dual ABCB1/ABCG2 inhibitor elacridar. Sunitinib 213-222 ATP-binding cassette, sub-family B (MDR/TAP), member 1B Mus musculus 95-100 21351087-2 2012 We aimed to investigate the in vivo roles of the ATP-binding cassette drug efflux transporters ABCB1 and ABCG2 in plasma pharmacokinetics and brain accumulation of oral sunitinib, and the feasibility of improving sunitinib kinetics using oral coadministration of the dual ABCB1/ABCG2 inhibitor elacridar. Sunitinib 213-222 ATP binding cassette subfamily G member 2 (Junior blood group) Mus musculus 105-110 21351087-2 2012 We aimed to investigate the in vivo roles of the ATP-binding cassette drug efflux transporters ABCB1 and ABCG2 in plasma pharmacokinetics and brain accumulation of oral sunitinib, and the feasibility of improving sunitinib kinetics using oral coadministration of the dual ABCB1/ABCG2 inhibitor elacridar. Sunitinib 213-222 ATP-binding cassette, sub-family B (MDR/TAP), member 1B Mus musculus 272-277 21351087-2 2012 We aimed to investigate the in vivo roles of the ATP-binding cassette drug efflux transporters ABCB1 and ABCG2 in plasma pharmacokinetics and brain accumulation of oral sunitinib, and the feasibility of improving sunitinib kinetics using oral coadministration of the dual ABCB1/ABCG2 inhibitor elacridar. Sunitinib 213-222 ATP binding cassette subfamily G member 2 (Junior blood group) Mus musculus 278-283 21351087-4 2012 In vitro, sunitinib was a good substrate of murine (mu)ABCG2 and a moderate substrate of human (hu)ABCB1 and huABCG2. Sunitinib 10-19 ATP binding cassette subfamily G member 2 (Junior blood group) Mus musculus 55-60 21351087-4 2012 In vitro, sunitinib was a good substrate of murine (mu)ABCG2 and a moderate substrate of human (hu)ABCB1 and huABCG2. Sunitinib 10-19 ATP binding cassette subfamily B member 1 Homo sapiens 99-104 21351087-6 2012 Brain accumulation of sunitinib was markedly (23-fold) increased in Abcb1a/b/Abcg2(-/-) mice, but only slightly (2.3-fold) in Abcb1a/b(-/-) mice, and not in Abcg2(-/-) mice. Sunitinib 22-31 ATP-binding cassette, sub-family B (MDR/TAP), member 1A Mus musculus 68-74 21351087-6 2012 Brain accumulation of sunitinib was markedly (23-fold) increased in Abcb1a/b/Abcg2(-/-) mice, but only slightly (2.3-fold) in Abcb1a/b(-/-) mice, and not in Abcg2(-/-) mice. Sunitinib 22-31 ATP binding cassette subfamily G member 2 (Junior blood group) Mus musculus 77-82 21351087-6 2012 Brain accumulation of sunitinib was markedly (23-fold) increased in Abcb1a/b/Abcg2(-/-) mice, but only slightly (2.3-fold) in Abcb1a/b(-/-) mice, and not in Abcg2(-/-) mice. Sunitinib 22-31 ATP binding cassette subfamily G member 2 (Junior blood group) Mus musculus 159-164 21351087-7 2012 Importantly, a clinically realistic coadministration of oral elacridar and oral sunitinib to wild-type mice resulted in markedly increased sunitinib brain accumulation, equaling levels in Abcb1a/1b/Abcg2(-/-) mice. Sunitinib 80-89 ATP-binding cassette, sub-family B (MDR/TAP), member 1A Mus musculus 188-194 21351087-7 2012 Importantly, a clinically realistic coadministration of oral elacridar and oral sunitinib to wild-type mice resulted in markedly increased sunitinib brain accumulation, equaling levels in Abcb1a/1b/Abcg2(-/-) mice. Sunitinib 80-89 ATP binding cassette subfamily G member 2 (Junior blood group) Mus musculus 198-203 21876693-4 2012 Research on angiogenesis in general, and vascular endothelial growth factor in particular, is a major focus in biomedicine and has led to the clinical approval of several antiangiogenic agents including thalidomide, bevacizumab, sorafenib, sunitinib, pazopanib, temesirolimus, and everolimus. Sunitinib 240-249 vascular endothelial growth factor A Homo sapiens 41-75 21961001-3 2012 On this basis, agents rendering VEGF ineffective by neutralizing VEGF (bevacizumab), blocking its receptors (aflibercept), or interfering with the postreceptor signaling pathways (sunitinib) provide us with the rational treatment options. Sunitinib 180-189 vascular endothelial growth factor A Homo sapiens 32-36 22357730-5 2012 Specifically, phase III studies indicate that pharmacologic inhibition of the vascular endothelial growth factor pathway with sunitinib, and of the mammalian target of rapamycin pathway with everolimus, appears to have altered the natural history of these diseases. Sunitinib 126-135 vascular endothelial growth factor A Homo sapiens 78-112 21880693-5 2012 Binding analysis of the inhibitors to the two different phosphorylation forms of CSF-1R, using surface plasmon resonance spectrometry, revealed that staurosporine bound to both forms with similar affinity, but sunitinib bound to the dephosphorylated form with higher affinity. Sunitinib 210-219 colony stimulating factor 1 receptor Homo sapiens 81-87 21880693-6 2012 Thus, these observations suggest that sunitinib binds preferentially to the inactive form, preventing the activation of CSF-1R. Sunitinib 38-47 colony stimulating factor 1 receptor Homo sapiens 120-126 22677881-6 2012 In second-line treatment, sunitinib showed a significantly longer PFS2 than sorafenib (average increase 2.66 months, 95% CI 1.02-4.3, p = 0.003). Sunitinib 26-35 GINS complex subunit 2 Homo sapiens 66-70 23028796-7 2012 Sunitinib could induce more apoptotic cells in 786-0 cells with knockdown EpoR expression. Sunitinib 0-9 erythropoietin receptor Homo sapiens 74-78 23056179-8 2012 However, the in vivo efficacy testing of the gefitinib and sunitinib combination in an EGFR amplified/PTEN wild type GBM xenograft model revealed that gefitinib alone could significantly improve survival in animals whereas sunitinib did not show any survival benefit. Sunitinib 59-68 epidermal growth factor receptor Homo sapiens 87-91 23056179-8 2012 However, the in vivo efficacy testing of the gefitinib and sunitinib combination in an EGFR amplified/PTEN wild type GBM xenograft model revealed that gefitinib alone could significantly improve survival in animals whereas sunitinib did not show any survival benefit. Sunitinib 59-68 phosphatase and tensin homolog Homo sapiens 102-106 23028796-8 2012 Our results suggested that Epo/EpoR pathway was involved in cell growth, invasion, survival, and sensitivity to the multi-kinases inhibitor Sunitinib in RCC cells. Sunitinib 140-149 erythropoietin Homo sapiens 27-30 23028796-8 2012 Our results suggested that Epo/EpoR pathway was involved in cell growth, invasion, survival, and sensitivity to the multi-kinases inhibitor Sunitinib in RCC cells. Sunitinib 140-149 erythropoietin receptor Homo sapiens 31-35 22723862-6 2012 In endothelial HUVECs, gemcitabine, bevacizumab, sunitinib and EMAP caused 70, 41, 86 and 67 percent inhibition, while combination of gemcitabine with bevacizumab, sunitinib or EMAP had additive effects. Sunitinib 49-58 EMAP like 1 Homo sapiens 177-181 22723862-6 2012 In endothelial HUVECs, gemcitabine, bevacizumab, sunitinib and EMAP caused 70, 41, 86 and 67 percent inhibition, while combination of gemcitabine with bevacizumab, sunitinib or EMAP had additive effects. Sunitinib 164-173 EMAP like 1 Homo sapiens 63-67 22412884-9 2012 The marginal cost-effectiveness (cost per additional QALY) gained via the sunitinib strategy compared with the conventional strategy was $220,384 (without SPAP, interleukin-2 plus interferon-alfa and bevacizumab plus interferon-alfa were dominated) and $16,993 (with SPAP, interferon-alfa, interleukin-2 plus interferon-alfa and bevacizumab plus interferon-alfa were dominated). Sunitinib 74-83 interleukin 2 Homo sapiens 290-303 22662219-7 2012 P53 reactivation substantially improved the apoptotic response after effective KIT inhibition with sunitinib and 17-AAG in IM-resistant cell lines. Sunitinib 99-108 tumor protein p53 Homo sapiens 0-3 22745791-3 2012 We investigated the renal effects of the administration, during 45 days, of sunitinib (Su), a VEGF receptor inhibitor, to rats with 5/6 renal ablation (Nx). Sunitinib 76-85 vascular endothelial growth factor A Rattus norvegicus 94-98 22229044-1 2012 Based on preclinical data available in the RIP1-Tag2 transgenic mouse model, sunitinib is an inhibitor of angiogenesis in pancreatic neuroendocrine tumors blocking vascular endothelial growth factor receptors and platelet-derived growth factor receptors in endothelial cells and pericytes, respectively. Sunitinib 77-86 receptor (TNFRSF)-interacting serine-threonine kinase 1 Mus musculus 43-47 22084065-3 2011 We evaluated sunitinib, an angiogenesis inhibitor that targets VEGF and PDGF receptor signaling, in two GEMMs of pancreatic cancer. Sunitinib 13-22 vascular endothelial growth factor A Mus musculus 63-67 23650573-0 2012 IGF-1 receptor is down-regulated by sunitinib induces MDM2-dependent ubiquitination. Sunitinib 36-45 insulin like growth factor 1 Homo sapiens 0-5 23650573-2 2012 The aim is to investigate the effects of sunitinib on IGF-1R cell signaling transduction, especially on receptor phosphorylation and ubiquitination. Sunitinib 41-50 insulin like growth factor 1 receptor Homo sapiens 54-60 23650573-4 2012 However, the phosphorylations of receptor tyrosine, Akt and ERK were significant inhibited by sunitinib. Sunitinib 94-103 AKT serine/threonine kinase 1 Homo sapiens 52-55 23650573-4 2012 However, the phosphorylations of receptor tyrosine, Akt and ERK were significant inhibited by sunitinib. Sunitinib 94-103 mitogen-activated protein kinase 1 Homo sapiens 60-63 23650573-5 2012 We found that both IGF-1 and sunitinib obviously down regulated the IGF-1R expression. Sunitinib 29-38 insulin like growth factor 1 receptor Homo sapiens 68-74 23650573-6 2012 For analysis the ubiquitination, HEK293 cells were simulated with 100 ng/ml IGF-1 or 10 nM sunitinib for 10 min after serum starvation for 24 h. Both IGF-1 and sunitinib could obviously induce the IGF-1R ubiquitination at 10 min compared with control (only serum free, no stimulation), indicating IGF-1 and sunitinib down-regulate the IGF-1R by increasing the receptor degradation through ubiquitination dependent proteasome pathway. Sunitinib 91-100 insulin like growth factor 1 Homo sapiens 150-155 23650573-6 2012 For analysis the ubiquitination, HEK293 cells were simulated with 100 ng/ml IGF-1 or 10 nM sunitinib for 10 min after serum starvation for 24 h. Both IGF-1 and sunitinib could obviously induce the IGF-1R ubiquitination at 10 min compared with control (only serum free, no stimulation), indicating IGF-1 and sunitinib down-regulate the IGF-1R by increasing the receptor degradation through ubiquitination dependent proteasome pathway. Sunitinib 91-100 insulin like growth factor 1 receptor Homo sapiens 197-203 23650573-6 2012 For analysis the ubiquitination, HEK293 cells were simulated with 100 ng/ml IGF-1 or 10 nM sunitinib for 10 min after serum starvation for 24 h. Both IGF-1 and sunitinib could obviously induce the IGF-1R ubiquitination at 10 min compared with control (only serum free, no stimulation), indicating IGF-1 and sunitinib down-regulate the IGF-1R by increasing the receptor degradation through ubiquitination dependent proteasome pathway. Sunitinib 91-100 insulin like growth factor 1 Homo sapiens 150-169 23650573-6 2012 For analysis the ubiquitination, HEK293 cells were simulated with 100 ng/ml IGF-1 or 10 nM sunitinib for 10 min after serum starvation for 24 h. Both IGF-1 and sunitinib could obviously induce the IGF-1R ubiquitination at 10 min compared with control (only serum free, no stimulation), indicating IGF-1 and sunitinib down-regulate the IGF-1R by increasing the receptor degradation through ubiquitination dependent proteasome pathway. Sunitinib 91-100 insulin like growth factor 1 receptor Homo sapiens 335-341 23650573-6 2012 For analysis the ubiquitination, HEK293 cells were simulated with 100 ng/ml IGF-1 or 10 nM sunitinib for 10 min after serum starvation for 24 h. Both IGF-1 and sunitinib could obviously induce the IGF-1R ubiquitination at 10 min compared with control (only serum free, no stimulation), indicating IGF-1 and sunitinib down-regulate the IGF-1R by increasing the receptor degradation through ubiquitination dependent proteasome pathway. Sunitinib 160-169 insulin like growth factor 1 Homo sapiens 76-81 23650573-6 2012 For analysis the ubiquitination, HEK293 cells were simulated with 100 ng/ml IGF-1 or 10 nM sunitinib for 10 min after serum starvation for 24 h. Both IGF-1 and sunitinib could obviously induce the IGF-1R ubiquitination at 10 min compared with control (only serum free, no stimulation), indicating IGF-1 and sunitinib down-regulate the IGF-1R by increasing the receptor degradation through ubiquitination dependent proteasome pathway. Sunitinib 160-169 insulin like growth factor 1 receptor Homo sapiens 197-203 23650573-6 2012 For analysis the ubiquitination, HEK293 cells were simulated with 100 ng/ml IGF-1 or 10 nM sunitinib for 10 min after serum starvation for 24 h. Both IGF-1 and sunitinib could obviously induce the IGF-1R ubiquitination at 10 min compared with control (only serum free, no stimulation), indicating IGF-1 and sunitinib down-regulate the IGF-1R by increasing the receptor degradation through ubiquitination dependent proteasome pathway. Sunitinib 160-169 insulin like growth factor 1 receptor Homo sapiens 335-341 23650573-7 2012 We also found that MDM2 combined to IGF-1R in response to sunitinib stimulation. Sunitinib 58-67 MDM2 proto-oncogene Homo sapiens 19-23 23650573-7 2012 We also found that MDM2 combined to IGF-1R in response to sunitinib stimulation. Sunitinib 58-67 insulin like growth factor 1 receptor Homo sapiens 36-42 23650573-11 2012 These results mean that sunitinib mediates ubiquitination of IGF-1R dependent on MDM2. Sunitinib 24-33 insulin like growth factor 1 receptor Homo sapiens 61-67 23650573-11 2012 These results mean that sunitinib mediates ubiquitination of IGF-1R dependent on MDM2. Sunitinib 24-33 MDM2 proto-oncogene Homo sapiens 81-85 23650573-12 2012 In summary, sunitinib could block signaling transduction and mediate degradation of IGF-1R. Sunitinib 12-21 insulin like growth factor 1 receptor Homo sapiens 84-90 22038997-10 2011 Sunitinib PK and VEGF ligand levels increased during sunitinib exposure and returned toward baseline during the treatment withdrawal. Sunitinib 53-62 vascular endothelial growth factor A Homo sapiens 17-21 22038997-11 2011 CONCLUSIONS: The increase of cellular proliferation during sunitinib withdrawal in patients with renal cell carcinoma and other solid malignancies is consistent with a VEGF receptor (VEGFR) tyrosine kinase inhibitor (TKI) withdrawal flare. Sunitinib 59-68 kinase insert domain receptor Homo sapiens 168-181 22038997-11 2011 CONCLUSIONS: The increase of cellular proliferation during sunitinib withdrawal in patients with renal cell carcinoma and other solid malignancies is consistent with a VEGF receptor (VEGFR) tyrosine kinase inhibitor (TKI) withdrawal flare. Sunitinib 59-68 kinase insert domain receptor Homo sapiens 183-188 22014215-11 2011 In addition, Zt/c9-Dox-IL in combination with small molecule inhibitors lapatinib, sunitinib, or dasatinib further reduced the viability of CSCs(+24/44/ESA). Sunitinib 83-92 paraoxonase 1 Homo sapiens 140-156 21415240-4 2011 RESULTS: Sunitinib demonstrated comparable dose-dependent growth inhibition in cisplatin-sensitive and cisplatin-resistant cell lines, with IC(50) between 3.0 and 3.8 muM. Sunitinib 9-18 latexin Homo sapiens 167-170 21826469-5 2011 This review discusses the relevant translational data from animal models of heart failure, focusing on three key pathways that are inhibited by sunitinib: AMP-activated protein kinase (AMPK), platelet-derived growth factor receptors (PDGFRs), and the vascular endothelial growth factor receptors (VEGFRs) 1, 2, and 3. Sunitinib 144-153 fms related receptor tyrosine kinase 1 Homo sapiens 251-316 22199315-2 2011 We investigated the IDH1 gene mutation status by nested PCR and denaturing gradient gel electrophoresis (DGGE) on DNA extracted from archival tumor blocks of 63 glioma patients who were treated following recurrence with the epidermal growth factor receptor (EGFR)-targeted blocking monoclonal antibody cetuximab, or the vascular endothelial growth factor (receptor) (VEGF(R))-targeted agents sunitinib malate and bevacizumab. Sunitinib 392-408 isocitrate dehydrogenase (NADP(+)) 1 Homo sapiens 20-24 22199315-4 2011 Patients with IDH1 mutation also had a superior OS from the time of recurrence when treated with sunitinib or bevacizumab but a worse OS when treated with cetuximab. Sunitinib 97-106 isocitrate dehydrogenase (NADP(+)) 1 Homo sapiens 14-18 21931979-4 2011 Initially developed for its inhibition of the vascular endothelial growth factor (VEGF) signaling pathway, sunitinib has been associated with hypertension and heart failure. Sunitinib 107-116 vascular endothelial growth factor A Homo sapiens 46-80 21931979-4 2011 Initially developed for its inhibition of the vascular endothelial growth factor (VEGF) signaling pathway, sunitinib has been associated with hypertension and heart failure. Sunitinib 107-116 vascular endothelial growth factor A Homo sapiens 82-86 20467883-1 2011 PURPOSE: Sunitinib is a multi-target receptor tyrosine kinase (RTK) inhibitor against vascular endothelial growth factor receptors, platelet-derived growth factor receptors (PDGFR), c-kit and RET. Sunitinib 9-18 TYRO3 protein tyrosine kinase 3 Mus musculus 37-61 22098229-2 2011 Currently available oral multitargeted VEGF tyrosine kinase inhibitors (TKIs) that have been approved by the US Food and Drug Administration for advanced RCC, include sunitinib, sorafenib and pazopanib. Sunitinib 167-176 vascular endothelial growth factor A Homo sapiens 39-43 20676744-0 2011 VEGF pathway inhibition by anticancer agent sunitinib and susceptibility to atherosclerosis plaque disruption. Sunitinib 44-53 vascular endothelial growth factor A Homo sapiens 0-4 20676744-3 2011 We report on a case of bowel infarction in a renal cancer patient treated with the anti-VEGF agent sunitinib. Sunitinib 99-108 vascular endothelial growth factor A Homo sapiens 88-92 20467883-1 2011 PURPOSE: Sunitinib is a multi-target receptor tyrosine kinase (RTK) inhibitor against vascular endothelial growth factor receptors, platelet-derived growth factor receptors (PDGFR), c-kit and RET. Sunitinib 9-18 TYRO3 protein tyrosine kinase 3 Mus musculus 63-66 20467883-1 2011 PURPOSE: Sunitinib is a multi-target receptor tyrosine kinase (RTK) inhibitor against vascular endothelial growth factor receptors, platelet-derived growth factor receptors (PDGFR), c-kit and RET. Sunitinib 9-18 platelet derived growth factor receptor, beta polypeptide Mus musculus 132-172 20467883-1 2011 PURPOSE: Sunitinib is a multi-target receptor tyrosine kinase (RTK) inhibitor against vascular endothelial growth factor receptors, platelet-derived growth factor receptors (PDGFR), c-kit and RET. Sunitinib 9-18 platelet derived growth factor receptor, beta polypeptide Mus musculus 174-179 20467883-1 2011 PURPOSE: Sunitinib is a multi-target receptor tyrosine kinase (RTK) inhibitor against vascular endothelial growth factor receptors, platelet-derived growth factor receptors (PDGFR), c-kit and RET. Sunitinib 9-18 KIT proto-oncogene receptor tyrosine kinase Mus musculus 182-187 20467883-1 2011 PURPOSE: Sunitinib is a multi-target receptor tyrosine kinase (RTK) inhibitor against vascular endothelial growth factor receptors, platelet-derived growth factor receptors (PDGFR), c-kit and RET. Sunitinib 9-18 ret proto-oncogene Mus musculus 192-195 22027642-6 2011 Sunitinib is a multikinase inhibitor that targets the PDGF-alpha- and ?beta-, VEGF-1-3-, KIT-, FLT3-, CSF-1- and RET-receptor, thereby impairing tumour proliferation, pathological angiogenesis and metastasation. Sunitinib 0-9 fms related receptor tyrosine kinase 3 Homo sapiens 95-99 21906609-2 2011 Although the precise mechanism of sunitinib cardiotoxicity is not known, both the key metabolic energy regulator, AMP-activated protein kinase (AMPK), and ribosomal S 6 kinase (RSK) have been hypothesized as causative, albeit based on rodent models. Sunitinib 34-43 ribosomal protein S6 kinase A2 Homo sapiens 177-180 21744079-1 2011 We determined the maximum tolerated dose (MTD) and dose-limiting toxicities (DLT) of the oral vascular endothelial growth factor receptor (VEGFR) inhibitor, sunitinib, when administered with irinotecan among recurrent malignant glioma (MG) patients. Sunitinib 157-166 kinase insert domain receptor Homo sapiens 94-137 21744079-1 2011 We determined the maximum tolerated dose (MTD) and dose-limiting toxicities (DLT) of the oral vascular endothelial growth factor receptor (VEGFR) inhibitor, sunitinib, when administered with irinotecan among recurrent malignant glioma (MG) patients. Sunitinib 157-166 kinase insert domain receptor Homo sapiens 139-144 22027642-6 2011 Sunitinib is a multikinase inhibitor that targets the PDGF-alpha- and ?beta-, VEGF-1-3-, KIT-, FLT3-, CSF-1- and RET-receptor, thereby impairing tumour proliferation, pathological angiogenesis and metastasation. Sunitinib 0-9 colony stimulating factor 1 Homo sapiens 102-125 21954449-12 2011 DISCUSSION: Sunitinib is a small molecule that inhibits multiple receptor tyrosine kinases such as stem cell factor receptor, vascular endothelial growth factor, and platelet-derived growth factor. Sunitinib 12-21 vascular endothelial growth factor A Homo sapiens 126-160 22015557-6 2011 SGI-1776 in combination with sunitinib induced a further reduction in c-Myc levels, which was associated with enhanced anticancer activity. Sunitinib 29-38 MYC proto-oncogene, bHLH transcription factor Homo sapiens 70-75 22015557-7 2011 siRNA-mediated knockdown of c-Myc demonstrated that its expression has a key role in regulating the sensitivity to the combination of SGI-1776 and sunitinib. Sunitinib 147-156 MYC proto-oncogene, bHLH transcription factor Homo sapiens 28-33 21952069-18 2011 A preliminary report of the investigational VEGF receptorinhibitor axitinib gave superior PFS to sorafenib after either prior cytokine or prior sunitinib treatment. Sunitinib 144-153 vascular endothelial growth factor A Homo sapiens 44-48 21933109-1 2011 Research on the formation of new blood vessels (angiogenesis) in general and vascular endothelial growth factor (VEGF) in particular is a major focus in biomedicine and has led to the clinical approval of the monoclonal anti- VEGF antibody bevazicumab; and the second-generation multitargeted receptor kinase inhibitors (RTKIs) sorafenib, sunitinib, and pazopanib. Sunitinib 339-348 vascular endothelial growth factor A Homo sapiens 226-230 22166826-7 2011 The chemotherapy (gemcitabine and cisplatin) and the molecular target therapy (sunitinib) were administerd but the primary lesion and metastases was progressive and serum G-CSF concentration was elevated to 229 pg/ml. Sunitinib 79-88 colony stimulating factor 3 Homo sapiens 171-176 21965162-1 2011 We report the case of a patient with metastatic renal cell carcinoma with Xp11.2 translocation/transcription factor E3 (TFE3) gene fusion who had presented with sunitinib-induced nephrotic syndrome in association with favorable and durable treatment response. Sunitinib 161-170 transcription factor binding to IGHM enhancer 3 Homo sapiens 81-118 21965162-1 2011 We report the case of a patient with metastatic renal cell carcinoma with Xp11.2 translocation/transcription factor E3 (TFE3) gene fusion who had presented with sunitinib-induced nephrotic syndrome in association with favorable and durable treatment response. Sunitinib 161-170 transcription factor binding to IGHM enhancer 3 Homo sapiens 120-124 22029859-5 2011 This correlated with viral NS1 expression levels and was enhanced by combination with chemotherapeutic agents or sunitinib. Sunitinib 113-122 influenza virus NS1A binding protein Homo sapiens 27-30 21883349-4 2011 Recently, sunitinib, a multityrosine kinase inhibitor (TKI), has been found to be rapidly metabolized by CYP3A4 during mitotane treatment, indicating clinically relevant drug interactions with mitotane. Sunitinib 10-19 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 105-111 22005473-5 2011 METHODS: This open-labeled phase II trial evaluated single-agent sunitinib, an inhibitor of multiple receptor tyrosine kinases including the vascular endothelial growth factor receptors, given at 50 mg daily orally for 4 weeks followed by a 2-week rest, in patients with advanced MPM. Sunitinib 65-74 vascular endothelial growth factor A Homo sapiens 141-175 22015057-10 2011 Two VEGFR3 missense polymorphisms were associated with reduced PFS with sunitinib on multivariable analysis: rs307826 (hazard ratio [HR] per allele 3 57, 1 75-7 30; p(unadjusted)=0 00049, p(adjusted)=0 0079) and rs307821 (3 31, 1 64-6 68; p(unadjusted)=0 00085, p(adjusted)=0 014). Sunitinib 72-81 fms related receptor tyrosine kinase 4 Homo sapiens 4-10 22015057-13 2011 INTERPRETATION: Polymorphisms in VEGFR3 and CYP3A5*1 might be able to define a subset of patients with renal-cell carcinoma with decreased sunitinib response and tolerability. Sunitinib 139-148 fms related receptor tyrosine kinase 4 Homo sapiens 33-39 22015057-13 2011 INTERPRETATION: Polymorphisms in VEGFR3 and CYP3A5*1 might be able to define a subset of patients with renal-cell carcinoma with decreased sunitinib response and tolerability. Sunitinib 139-148 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 44-50 21456006-9 2011 The objective response rate was 3%, comprising 1 PR in a patient with a GIST possessing both a KIT exon 11 mutation, and an imatinib-resistant and sunitinib-resistant KIT exon 17 mutation. Sunitinib 147-156 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 167-170 21951737-2 2011 Four agents antagonizing vascular endothelial growth factor-mediated signaling have been approved for the treatment of metastatic RCC, including the monoclonal antibody bevacizumab and the small molecular inhibitors sunitinib, sorafenib, and pazopanib. Sunitinib 216-225 vascular endothelial growth factor A Homo sapiens 25-59 21970485-2 2011 They harbor specific activating mutations in the KIT or platelet-derived growth factor receptor alpha ( PDGFRA ) receptor tyrosine kinases, which makes them responsive to pharmacologic inhibitors, such as imatinib mesylate and sunitinib malate. Sunitinib 227-243 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 49-52 21970485-2 2011 They harbor specific activating mutations in the KIT or platelet-derived growth factor receptor alpha ( PDGFRA ) receptor tyrosine kinases, which makes them responsive to pharmacologic inhibitors, such as imatinib mesylate and sunitinib malate. Sunitinib 227-243 platelet derived growth factor receptor alpha Homo sapiens 56-101 21970485-2 2011 They harbor specific activating mutations in the KIT or platelet-derived growth factor receptor alpha ( PDGFRA ) receptor tyrosine kinases, which makes them responsive to pharmacologic inhibitors, such as imatinib mesylate and sunitinib malate. Sunitinib 227-243 platelet derived growth factor receptor alpha Homo sapiens 104-110 22654799-0 2011 Sunitinib Inhibits Cell Proliferation and Alters Steroidogenesis by Down-Regulation of HSD3B2 in Adrenocortical Carcinoma Cells. Sunitinib 0-9 hydroxy-delta-5-steroid dehydrogenase, 3 beta- and steroid delta-isomerase 2 Homo sapiens 87-93 22654799-14 2011 Sunitinib exhibits anti-proliferative effects in vitro, and appears to specifically block adrenal steroidogenesis by down-regulation of HSD3B2, rendering it a promising option for treatment of ACC. Sunitinib 0-9 hydroxy-delta-5-steroid dehydrogenase, 3 beta- and steroid delta-isomerase 2 Homo sapiens 136-142 21771900-6 2011 Treatment with the Flt3 inhibitor sunitinib was started together with mBSA immunization or together with the induction of arthritis. Sunitinib 34-43 fms related receptor tyrosine kinase 3 Homo sapiens 19-23 21128230-8 2011 The administration of sunitinib, a drug known to inhibit PDGFR, was associated in murine models and in cancer patients with an increase of apoptotic/necrotic circulating PPC, suggesting a direct targeting of these cells. Sunitinib 22-31 platelet derived growth factor receptor, beta polypeptide Mus musculus 57-62 22654799-11 2011 Sunitinib induced down-regulation of HSD3B2 mRNA and protein in ACC cell lines (mRNA: 1 muM 44 +- 16%*; 5 muM 22 +- 2%*; 10 muM 19 +- 4%*; protein: 1 muM 82 +- 8%; 5 muM 63 +- 8%*; 10 muM 55 +- 9%*). Sunitinib 0-9 hydroxy-delta-5-steroid dehydrogenase, 3 beta- and steroid delta-isomerase 2 Homo sapiens 37-43 21725210-0 2011 Sunitinib inhibits papillary thyroid carcinoma with RET/PTC rearrangement but not BRAF mutation. Sunitinib 0-9 ret proto-oncogene Homo sapiens 52-55 21265994-4 2011 RESULTS: Sunitinib was more effective and less costly than sorafenib (gains of 0.52 PFLYs, 0.16 LYs and 0.17 QALYs and savings/patient of $13,576 in the US) and bevacizumab plus IFN-alpha (gains of 0.19 PFLYs, 0.23 LYs and 0.16 QALYs in both countries and savings/patient of $67,798 and $47,264 in the US and Sweden, respectively). Sunitinib 11-20 interferon alpha 1 Homo sapiens 180-189 21725210-0 2011 Sunitinib inhibits papillary thyroid carcinoma with RET/PTC rearrangement but not BRAF mutation. Sunitinib 0-9 ret proto-oncogene Homo sapiens 56-59 21725210-4 2011 Cell growth of papillary thyroid cancer cells with RET/PTC rearrangement was effectively inhibited at low doses of sunitinib (IC50=0.658 muM), whereas that of BRAF mutated cells required higher doses. Sunitinib 115-124 ret proto-oncogene Homo sapiens 51-54 21725210-4 2011 Cell growth of papillary thyroid cancer cells with RET/PTC rearrangement was effectively inhibited at low doses of sunitinib (IC50=0.658 muM), whereas that of BRAF mutated cells required higher doses. Sunitinib 115-124 ret proto-oncogene Homo sapiens 55-58 21725210-7 2011 In vivo orthotopic thyroid cancer mouse model demonstrated statistically significant tumor growth inhibition by sunitinib in RET/PTC rearrangement cancer cells. Sunitinib 112-121 ret proto-oncogene Mus musculus 125-128 21737509-2 2011 Imatinib (IMA) and sunitinib (SUN) are very effective KIT-inhibitors in patients with advanced GIST but have no curative potential. Sunitinib 19-28 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 54-57 21725210-7 2011 In vivo orthotopic thyroid cancer mouse model demonstrated statistically significant tumor growth inhibition by sunitinib in RET/PTC rearrangement cancer cells. Sunitinib 112-121 ret proto-oncogene Mus musculus 129-132 21725210-8 2011 We conclude that sunitinib effectively inhibits RET/PTC rearrangement cells but not BRAF mutated cells. Sunitinib 17-26 ret proto-oncogene Homo sapiens 48-51 21725210-8 2011 We conclude that sunitinib effectively inhibits RET/PTC rearrangement cells but not BRAF mutated cells. Sunitinib 17-26 ret proto-oncogene Homo sapiens 52-55 21725210-9 2011 These data suggest that sunitinib exerts its effect by inhibiting the upstream MAPK signaling cascade. Sunitinib 24-33 mitogen-activated protein kinase 1 Homo sapiens 79-83 21796416-0 2011 Synergistic interaction between sunitinib and docetaxel is sequence dependent in human non-small lung cancer with EGFR TKIs-resistant mutation. Sunitinib 32-41 epidermal growth factor receptor Homo sapiens 114-118 21817899-0 2011 VEGF expression is related to good response and long progression-free survival in gastrointestinal stromal tumor patients treated with Sunitinib. Sunitinib 135-144 vascular endothelial growth factor A Homo sapiens 0-4 21817899-8 2011 In the sunitinib treatment, VEGF expression was related to a favorable response (P=0.002) and long PFS (P=0.020) in univariate analysis. Sunitinib 7-16 vascular endothelial growth factor A Homo sapiens 28-32 21817899-9 2011 CD34 (P=0.023) and PDGFR (P=0.022) expressions were also related to long sunitinib PFS in univariate analysis. Sunitinib 73-82 CD34 molecule Homo sapiens 0-4 21817899-9 2011 CD34 (P=0.023) and PDGFR (P=0.022) expressions were also related to long sunitinib PFS in univariate analysis. Sunitinib 73-82 platelet derived growth factor receptor beta Homo sapiens 19-24 21817899-11 2011 In conclusion, expressions of VEGF, PDGFR, and CD34 may have predictive value in sunitinib treatments. Sunitinib 81-90 vascular endothelial growth factor A Homo sapiens 30-34 21817899-11 2011 In conclusion, expressions of VEGF, PDGFR, and CD34 may have predictive value in sunitinib treatments. Sunitinib 81-90 platelet derived growth factor receptor beta Homo sapiens 36-41 21817899-11 2011 In conclusion, expressions of VEGF, PDGFR, and CD34 may have predictive value in sunitinib treatments. Sunitinib 81-90 CD34 molecule Homo sapiens 47-51 21843259-9 2011 Su increased apoptosis, as evidenced by the cleavage of caspase-3 and PARP-1 proteins. Sunitinib 0-2 caspase 3 Mus musculus 56-65 21843259-9 2011 Su increased apoptosis, as evidenced by the cleavage of caspase-3 and PARP-1 proteins. Sunitinib 0-2 poly (ADP-ribose) polymerase family, member 1 Mus musculus 70-76 22040503-1 2011 BACKGROUND: The tyrosine kinase inhibitors (TKIs) sunitinib, the first targeted agent for the first line treatment of metastatic renal cell carcinoma (RCC), targets the vascular endothelial growth factor (VEGF) pathway. Sunitinib 50-59 vascular endothelial growth factor A Homo sapiens 169-203 22040503-1 2011 BACKGROUND: The tyrosine kinase inhibitors (TKIs) sunitinib, the first targeted agent for the first line treatment of metastatic renal cell carcinoma (RCC), targets the vascular endothelial growth factor (VEGF) pathway. Sunitinib 50-59 vascular endothelial growth factor A Homo sapiens 205-209 21796416-10 2011 CONCLUSIONS: Sunitinib as a single agent exhibits anti-proliferative effects in vitro in NSCLC cell lines with EGFR T790M and K-ras mutations but the sequential administration of docetaxel followed by sunitinib is superior to sunitinib followed by docetaxel and concurrent administration. Sunitinib 13-22 epidermal growth factor receptor Homo sapiens 111-115 21796416-10 2011 CONCLUSIONS: Sunitinib as a single agent exhibits anti-proliferative effects in vitro in NSCLC cell lines with EGFR T790M and K-ras mutations but the sequential administration of docetaxel followed by sunitinib is superior to sunitinib followed by docetaxel and concurrent administration. Sunitinib 13-22 KRAS proto-oncogene, GTPase Homo sapiens 126-131 21508896-6 2011 Sunitinib, a kinase inhibitor, blocked several receptors, including vascular endothelial growth factor receptor 3 (VEGFR3), the major growth factor receptor for lymphatic endothelial cells. Sunitinib 0-9 FMS-like tyrosine kinase 4 Mus musculus 68-113 21316102-8 2011 In addition, both PD173074 and sunitinib inhibited the activation of FGFR3 in t(4;14)-positive MM cells. Sunitinib 31-40 fibroblast growth factor receptor 3 Homo sapiens 69-74 21316102-10 2011 CONCLUSIONS: These data demonstrate that PD173074 and sunitinib are inhibitors of FGFR3 in MM cell lines, and that sunitinib has in vivo activity in a human MM tumour xenograft model. Sunitinib 54-63 fibroblast growth factor receptor 3 Homo sapiens 82-87 21636578-0 2011 Inhibition of RNase L and RNA-dependent protein kinase (PKR) by sunitinib impairs antiviral innate immunity. Sunitinib 64-73 ribonuclease L (2', 5'-oligoisoadenylate synthetase-dependent) Mus musculus 14-21 21636578-0 2011 Inhibition of RNase L and RNA-dependent protein kinase (PKR) by sunitinib impairs antiviral innate immunity. Sunitinib 64-73 eukaryotic translation initiation factor 2-alpha kinase 2 Mus musculus 56-59 21636578-2 2011 Sunitinib is an orally available, ATP-competitive inhibitor of VEGF and PDGF receptors used clinically to suppress angiogenesis and tumor growth. Sunitinib 0-9 vascular endothelial growth factor A Mus musculus 63-67 21636578-3 2011 Sunitinib also impacts IRE1, an endoplasmic reticulum protein involved in the unfolded protein response that is closely related to RNase L. Sunitinib 0-9 endoplasmic reticulum (ER) to nucleus signalling 2 Mus musculus 23-27 21636578-3 2011 Sunitinib also impacts IRE1, an endoplasmic reticulum protein involved in the unfolded protein response that is closely related to RNase L. Sunitinib 0-9 ribonuclease L (2', 5'-oligoisoadenylate synthetase-dependent) Mus musculus 131-138 21636578-4 2011 Here, we report that sunitinib is a potent inhibitor of both RNase L and PKR with IC(50) values of 1.4 and 0.3 muM, respectively. Sunitinib 21-30 ribonuclease L (2', 5'-oligoisoadenylate synthetase-dependent) Mus musculus 61-68 21636578-4 2011 Here, we report that sunitinib is a potent inhibitor of both RNase L and PKR with IC(50) values of 1.4 and 0.3 muM, respectively. Sunitinib 21-30 eukaryotic translation initiation factor 2-alpha kinase 2 Mus musculus 73-76 21636578-8 2011 Furthermore, oral delivery of sunitinib in WT mice resulted in 10-fold higher viral titers in heart tissues while suppressing by about 2-fold the IFN-beta levels. Sunitinib 30-39 interferon beta 1, fibroblast Mus musculus 146-154 21636578-11 2011 Results indicate that sunitinib treatments prevent antiviral innate immune responses mediated by RNase L and PKR. Sunitinib 22-31 ribonuclease L (2', 5'-oligoisoadenylate synthetase-dependent) Mus musculus 97-104 21636578-11 2011 Results indicate that sunitinib treatments prevent antiviral innate immune responses mediated by RNase L and PKR. Sunitinib 22-31 eukaryotic translation initiation factor 2-alpha kinase 2 Mus musculus 109-112 21705501-2 2011 We report here 2 male patients with ETV6-FLT3(+) myeloid/lymphoid neoplasms with eosinophilia who were treated with the multitargeted TK inhibitors sunitinib and sorafenib. Sunitinib 148-157 ETS variant transcription factor 6 Homo sapiens 36-40 21705501-2 2011 We report here 2 male patients with ETV6-FLT3(+) myeloid/lymphoid neoplasms with eosinophilia who were treated with the multitargeted TK inhibitors sunitinib and sorafenib. Sunitinib 148-157 fms related receptor tyrosine kinase 3 Homo sapiens 41-45 21705501-2 2011 We report here 2 male patients with ETV6-FLT3(+) myeloid/lymphoid neoplasms with eosinophilia who were treated with the multitargeted TK inhibitors sunitinib and sorafenib. Sunitinib 148-157 TXK tyrosine kinase Homo sapiens 134-136 21787417-2 2011 Sunitinib has antiangiogenic activity and is an oral multitargeted inhibitor of vascular endothelial growth factor receptors (VEGFRs)-1, -2, and -3, platelet-derived growth factor receptors (PDGFRs)-alpha and -beta, stem-cell factor receptor (KIT), and other tyrosine kinases. Sunitinib 0-9 fms related receptor tyrosine kinase 1 Homo sapiens 80-147 21787417-2 2011 Sunitinib has antiangiogenic activity and is an oral multitargeted inhibitor of vascular endothelial growth factor receptors (VEGFRs)-1, -2, and -3, platelet-derived growth factor receptors (PDGFRs)-alpha and -beta, stem-cell factor receptor (KIT), and other tyrosine kinases. Sunitinib 0-9 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 243-246 21787417-11 2011 CONCLUSIONS: Baseline plasma VEGF-C levels predicted disease control (based on RECIST) and were positively associated with both TTP and OS in this exploratory analysis, suggesting that this VEGF family member may have utility in predicting clinical outcome in patients with HCC who receive sunitinib. Sunitinib 290-299 vascular endothelial growth factor C Homo sapiens 29-35 21787417-11 2011 CONCLUSIONS: Baseline plasma VEGF-C levels predicted disease control (based on RECIST) and were positively associated with both TTP and OS in this exploratory analysis, suggesting that this VEGF family member may have utility in predicting clinical outcome in patients with HCC who receive sunitinib. Sunitinib 290-299 vascular endothelial growth factor A Homo sapiens 29-33 21508896-6 2011 Sunitinib, a kinase inhibitor, blocked several receptors, including vascular endothelial growth factor receptor 3 (VEGFR3), the major growth factor receptor for lymphatic endothelial cells. Sunitinib 0-9 FMS-like tyrosine kinase 4 Mus musculus 115-121 21508896-6 2011 Sunitinib, a kinase inhibitor, blocked several receptors, including vascular endothelial growth factor receptor 3 (VEGFR3), the major growth factor receptor for lymphatic endothelial cells. Sunitinib 0-9 receptor-like tyrosine kinase Mus musculus 89-111 21242589-10 2011 Antiproliferative assays and biochemistry on short-term culture showed that sunitinib is able to markedly impair ASPS cells growth and switch-off PDGFRB. Sunitinib 76-85 platelet derived growth factor receptor beta Homo sapiens 146-152 21242589-13 2011 CONCLUSIONS: We confirm the clinical efficacy of sunitinib in ASPS, mediated by PDGFRB, VEGFR2, and RET, which are all expressed in tumor cells. Sunitinib 49-58 platelet derived growth factor receptor beta Homo sapiens 80-86 21242589-13 2011 CONCLUSIONS: We confirm the clinical efficacy of sunitinib in ASPS, mediated by PDGFRB, VEGFR2, and RET, which are all expressed in tumor cells. Sunitinib 49-58 kinase insert domain receptor Homo sapiens 88-94 21242589-13 2011 CONCLUSIONS: We confirm the clinical efficacy of sunitinib in ASPS, mediated by PDGFRB, VEGFR2, and RET, which are all expressed in tumor cells. Sunitinib 49-58 ret proto-oncogene Homo sapiens 100-103 21519258-4 2011 RECENT FINDINGS: The major mechanism of resistance toward imatinib and sunitinib is the development of secondary resistance mutations in the kinase domain of KIT. Sunitinib 71-80 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 158-161 21693010-0 2011 Sunitinib inhibits lymphatic endothelial cell functions and lymph node metastasis in a breast cancer model through inhibition of vascular endothelial growth factor receptor 3. Sunitinib 0-9 fms related receptor tyrosine kinase 4 Homo sapiens 129-174 21315783-3 2011 Sunitinib also targets myeloid derived suppressor cells (MDSCs) significantly reducing their accumulation in the peripheral blood and reversing T cell (IFNgamma) suppression in both mRCC patients and in murine tumor models. Sunitinib 0-9 interferon gamma Homo sapiens 152-160 21315783-9 2011 Additionally, in vitro and in vivo experiments showed that GM-CSF prolongs survival of MDSCs, thus protecting them from the effects of sunitinib via a pSTAT5-dependent pathway. Sunitinib 135-144 colony stimulating factor 2 Homo sapiens 59-65 21315783-15 2011 GM-CSF"s role in promoting MDSC survival in patient tumors is supported by the observation that GM-CSF is produced in short-term RCC cultures at levels capable of protecting MDSCs from sunitinib-induced cell death. Sunitinib 185-194 colony stimulating factor 2 Homo sapiens 0-6 21315783-15 2011 GM-CSF"s role in promoting MDSC survival in patient tumors is supported by the observation that GM-CSF is produced in short-term RCC cultures at levels capable of protecting MDSCs from sunitinib-induced cell death. Sunitinib 185-194 colony stimulating factor 2 Homo sapiens 96-102 20872043-2 2011 The KIT, PDGFR-alpha, and VEGFR2 genes are frequently amplified and expressed in HGG and are molecular targets for therapeutic inhibition by the small-molecule kinase inhibitor sunitinib malate. Sunitinib 177-193 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 4-7 20872043-2 2011 The KIT, PDGFR-alpha, and VEGFR2 genes are frequently amplified and expressed in HGG and are molecular targets for therapeutic inhibition by the small-molecule kinase inhibitor sunitinib malate. Sunitinib 177-193 platelet derived growth factor receptor alpha Homo sapiens 9-20 20872043-2 2011 The KIT, PDGFR-alpha, and VEGFR2 genes are frequently amplified and expressed in HGG and are molecular targets for therapeutic inhibition by the small-molecule kinase inhibitor sunitinib malate. Sunitinib 177-193 kinase insert domain receptor Homo sapiens 26-32 21577109-4 2011 Inhibition of CSF1R using small molecule inhibitors such as imatinib, nilotinib or sunitinib can result in clinical, radiological and functional improvement in the affected joint. Sunitinib 83-92 colony stimulating factor 1 receptor Homo sapiens 14-19 21772092-7 2011 Those patients who presented low serum VEGF showed prolonged progression-free survival when treated with sunitinib. Sunitinib 105-114 vascular endothelial growth factor A Homo sapiens 39-43 21693010-9 2011 Furthermore, sunitinib attenuated the cell-proliferation activity induced by VEGF-C/D and prevented VEGF-C-induced migration and tube formation of the LECs; however, anti-VEGFR2 treatment shows only a partial effect on the growth and functions of the LECs. Sunitinib 13-22 vascular endothelial growth factor C Homo sapiens 77-83 21693010-9 2011 Furthermore, sunitinib attenuated the cell-proliferation activity induced by VEGF-C/D and prevented VEGF-C-induced migration and tube formation of the LECs; however, anti-VEGFR2 treatment shows only a partial effect on the growth and functions of the LECs. Sunitinib 13-22 vascular endothelial growth factor C Homo sapiens 100-106 21693010-9 2011 Furthermore, sunitinib attenuated the cell-proliferation activity induced by VEGF-C/D and prevented VEGF-C-induced migration and tube formation of the LECs; however, anti-VEGFR2 treatment shows only a partial effect on the growth and functions of the LECs. Sunitinib 13-22 kinase insert domain receptor Homo sapiens 171-177 21693010-13 2011 This effect of sunitinib was more potent than that of DC101, an anti-mouse VEGFR-2 antibody. Sunitinib 15-24 kinase insert domain receptor Homo sapiens 75-82 21693010-14 2011 CONCLUSIONS: The results suggest that sunitinib might be beneficial for the treatment of breast cancer by suppressing lymphangiogenesis and lymph node metastasis, through inhibition, particularly important, of VEGFR-3. Sunitinib 38-47 fms related receptor tyrosine kinase 4 Homo sapiens 210-217 21693010-5 2011 The purpose of this study was to investigate the effects of sunitinib on vascular endothelial growth factor receptor 3 (VEGFR-3) and a related event, lymphangiogenesis. Sunitinib 60-69 fms related receptor tyrosine kinase 4 Homo sapiens 73-118 21693010-5 2011 The purpose of this study was to investigate the effects of sunitinib on vascular endothelial growth factor receptor 3 (VEGFR-3) and a related event, lymphangiogenesis. Sunitinib 60-69 fms related receptor tyrosine kinase 4 Homo sapiens 120-127 21693010-6 2011 METHODS: The effects of sunitinib on the degree of phosphorylation of VEGFR-2/3 and other signaling molecules was examined in lymphatic endothelial cells (LECs) treated with the drug; VEGF-induced LEC growth, migration, and tube formation were also examined. Sunitinib 24-33 kinase insert domain receptor Homo sapiens 70-79 21693010-6 2011 METHODS: The effects of sunitinib on the degree of phosphorylation of VEGFR-2/3 and other signaling molecules was examined in lymphatic endothelial cells (LECs) treated with the drug; VEGF-induced LEC growth, migration, and tube formation were also examined. Sunitinib 24-33 vascular endothelial growth factor A Homo sapiens 70-74 21693010-6 2011 METHODS: The effects of sunitinib on the degree of phosphorylation of VEGFR-2/3 and other signaling molecules was examined in lymphatic endothelial cells (LECs) treated with the drug; VEGF-induced LEC growth, migration, and tube formation were also examined. Sunitinib 24-33 C-C motif chemokine ligand 16 Homo sapiens 155-158 21693010-8 2011 RESULTS: First, in human LECs, sunitinib blocked both VEGFR-2 and VEGFR-3 phosphorylation induced by VEGF-C or VEGF-D, and abrogated the activation of the downstream molecules extracellular signal-regulated kinase 1/2 (ERK1/2) and Akt. Sunitinib 31-40 kinase insert domain receptor Homo sapiens 54-61 21693010-8 2011 RESULTS: First, in human LECs, sunitinib blocked both VEGFR-2 and VEGFR-3 phosphorylation induced by VEGF-C or VEGF-D, and abrogated the activation of the downstream molecules extracellular signal-regulated kinase 1/2 (ERK1/2) and Akt. Sunitinib 31-40 fms related receptor tyrosine kinase 4 Homo sapiens 66-73 21693010-8 2011 RESULTS: First, in human LECs, sunitinib blocked both VEGFR-2 and VEGFR-3 phosphorylation induced by VEGF-C or VEGF-D, and abrogated the activation of the downstream molecules extracellular signal-regulated kinase 1/2 (ERK1/2) and Akt. Sunitinib 31-40 vascular endothelial growth factor C Homo sapiens 101-107 21693010-8 2011 RESULTS: First, in human LECs, sunitinib blocked both VEGFR-2 and VEGFR-3 phosphorylation induced by VEGF-C or VEGF-D, and abrogated the activation of the downstream molecules extracellular signal-regulated kinase 1/2 (ERK1/2) and Akt. Sunitinib 31-40 vascular endothelial growth factor D Homo sapiens 111-117 21693010-8 2011 RESULTS: First, in human LECs, sunitinib blocked both VEGFR-2 and VEGFR-3 phosphorylation induced by VEGF-C or VEGF-D, and abrogated the activation of the downstream molecules extracellular signal-regulated kinase 1/2 (ERK1/2) and Akt. Sunitinib 31-40 mitogen-activated protein kinase 1 Homo sapiens 176-217 21693010-8 2011 RESULTS: First, in human LECs, sunitinib blocked both VEGFR-2 and VEGFR-3 phosphorylation induced by VEGF-C or VEGF-D, and abrogated the activation of the downstream molecules extracellular signal-regulated kinase 1/2 (ERK1/2) and Akt. Sunitinib 31-40 mitogen-activated protein kinase 3 Homo sapiens 219-225 21693010-8 2011 RESULTS: First, in human LECs, sunitinib blocked both VEGFR-2 and VEGFR-3 phosphorylation induced by VEGF-C or VEGF-D, and abrogated the activation of the downstream molecules extracellular signal-regulated kinase 1/2 (ERK1/2) and Akt. Sunitinib 31-40 AKT serine/threonine kinase 1 Homo sapiens 231-234 21704235-11 2011 An indirect comparison suggests that sunitinib is likely to demonstrate greater clinical benefit than sorafenib in terms of PFS (HR = 0.47; 95% CI, 0.316-0.713; P < 0.001), using IFN-alpha as the comparator. Sunitinib 37-46 interferon alpha 1 Homo sapiens 182-191 21672194-7 2011 A number of biologically targeted agents targeting the VEGF and mTOR signaling pathways have recently shown promise, with recent trials showing treatment with the VEGFR tyrosine kinase inhibitor sunitinib or the mTOR inhibitor everolimus improves progression-free survival in patients with advanced NET. Sunitinib 195-204 vascular endothelial growth factor A Homo sapiens 55-59 21672194-7 2011 A number of biologically targeted agents targeting the VEGF and mTOR signaling pathways have recently shown promise, with recent trials showing treatment with the VEGFR tyrosine kinase inhibitor sunitinib or the mTOR inhibitor everolimus improves progression-free survival in patients with advanced NET. Sunitinib 195-204 mechanistic target of rapamycin kinase Homo sapiens 64-68 21481584-2 2011 Everolimus is an orally administered inhibitor of the mammalian target of rapamycin that recently received approval from the European Medicines Agency for the treatment of advanced RCC that has progressed on or after treatment with vascular endothelial growth factor (VEGF)-targeted therapy, and from the United States Food and Drug Administration for treatment of advanced RCC after failure of sorafenib or sunitinib. Sunitinib 408-417 vascular endothelial growth factor A Homo sapiens 268-272 21334201-10 2011 The influence of Imatinib and Sunitinib therapy in metastasized GISTs with wild type genotype and c-kit exon 9 mutations needs further investigation. Sunitinib 30-39 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 98-103 21566429-9 2011 More recently, sunitinib, a new KIT/PDGFRA kinase inhibitor, has been tested in patients with GIST resistant to imatinib with promising results. Sunitinib 15-24 platelet derived growth factor receptor alpha Homo sapiens 36-42 21677482-4 2011 Sunitinib malate, a multi-targeted tyrosine kinase inhibitor that shows activity against KIT and other receptor tyrosine kinases, including PDGFR and vascular endothelial growth factor receptor, is the only treatment for imatinib-resistant GISTs that is covered by national health insurance in Japan as ofthis writing. Sunitinib 0-16 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 89-92 21677482-4 2011 Sunitinib malate, a multi-targeted tyrosine kinase inhibitor that shows activity against KIT and other receptor tyrosine kinases, including PDGFR and vascular endothelial growth factor receptor, is the only treatment for imatinib-resistant GISTs that is covered by national health insurance in Japan as ofthis writing. Sunitinib 0-16 platelet derived growth factor receptor beta Homo sapiens 140-145 21527770-1 2011 BACKGROUND: Hypertension (HTN) is an on-target effect of the vascular endothelial growth factor pathway inhibitor, sunitinib. Sunitinib 115-124 vascular endothelial growth factor A Homo sapiens 61-95 21266595-6 2011 In contrast, midazolam 1"-hydroxylation catalyzed by recombinant CYP3A5 was enhanced by the coexistence of sorafenib or sunitinib in a concentration-dependent manner, with saturation occurring at approximately 10 muM. Sunitinib 120-129 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 65-71 21266595-6 2011 In contrast, midazolam 1"-hydroxylation catalyzed by recombinant CYP3A5 was enhanced by the coexistence of sorafenib or sunitinib in a concentration-dependent manner, with saturation occurring at approximately 10 muM. Sunitinib 120-129 latexin Homo sapiens 213-216 21266595-8 2011 Sorafenib N-oxidation and sunitinib N-deethylation, the primary routes of metabolism, were predominantly catalyzed by CYP3A4 but not by CYP3A5. Sunitinib 26-35 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 118-124 21266595-11 2011 Our results thus showed that sorafenib and sunitinib activated midazolam 1"-hydroxylation by CYP3A5 but inhibited that by CYP3A4. Sunitinib 43-52 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 93-99 21266595-11 2011 Our results thus showed that sorafenib and sunitinib activated midazolam 1"-hydroxylation by CYP3A5 but inhibited that by CYP3A4. Sunitinib 43-52 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 122-128 21266595-12 2011 Unexpected drug interactions involving sorafenib and sunitinib might occur via heterotropic cooperativity of CYP3A5. Sunitinib 53-62 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 109-115 21584715-2 2011 Small molecule tyrosine kinase inhibitors such as sorafenib, sunitinib, and pazopanib inhibit the downstream pathways of all three of the vascular endothelial growth factor receptors (VEGFR 1, 2, and 3). Sunitinib 61-70 fms related receptor tyrosine kinase 1 Homo sapiens 184-201 21480952-1 2011 AIMS: Reversible posterior leucoencephalopathy syndrome (RPLS) has been reported following the use of anti-vascular endothelial growth factor (VEGF) agents such as bevacizumab, sorafinib and sunitinib. Sunitinib 191-200 vascular endothelial growth factor A Homo sapiens 102-141 21480952-1 2011 AIMS: Reversible posterior leucoencephalopathy syndrome (RPLS) has been reported following the use of anti-vascular endothelial growth factor (VEGF) agents such as bevacizumab, sorafinib and sunitinib. Sunitinib 191-200 vascular endothelial growth factor A Homo sapiens 143-147 21469976-2 2011 Sunitinib is an oral multitargeted inhibitor of vascular endothelial growth factor receptors (VEGFRs-1, -2, and -3), platelet-derived growth factor receptors (PDGFRs-alpha and -beta), stem-cell factor receptor (KIT), FMS-like tyrosine kinase 3 (FLT3), colony-stimulating factor 1 receptor (CSF-1R), and glial cell line-derived neurotrophic factor receptor (REarranged during Transfection; RET). Sunitinib 0-9 fms related receptor tyrosine kinase 1 Homo sapiens 94-114 21469976-2 2011 Sunitinib is an oral multitargeted inhibitor of vascular endothelial growth factor receptors (VEGFRs-1, -2, and -3), platelet-derived growth factor receptors (PDGFRs-alpha and -beta), stem-cell factor receptor (KIT), FMS-like tyrosine kinase 3 (FLT3), colony-stimulating factor 1 receptor (CSF-1R), and glial cell line-derived neurotrophic factor receptor (REarranged during Transfection; RET). Sunitinib 0-9 platelet derived growth factor receptor alpha Homo sapiens 159-181 21469976-2 2011 Sunitinib is an oral multitargeted inhibitor of vascular endothelial growth factor receptors (VEGFRs-1, -2, and -3), platelet-derived growth factor receptors (PDGFRs-alpha and -beta), stem-cell factor receptor (KIT), FMS-like tyrosine kinase 3 (FLT3), colony-stimulating factor 1 receptor (CSF-1R), and glial cell line-derived neurotrophic factor receptor (REarranged during Transfection; RET). Sunitinib 0-9 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 211-214 21469976-2 2011 Sunitinib is an oral multitargeted inhibitor of vascular endothelial growth factor receptors (VEGFRs-1, -2, and -3), platelet-derived growth factor receptors (PDGFRs-alpha and -beta), stem-cell factor receptor (KIT), FMS-like tyrosine kinase 3 (FLT3), colony-stimulating factor 1 receptor (CSF-1R), and glial cell line-derived neurotrophic factor receptor (REarranged during Transfection; RET). Sunitinib 0-9 fms related receptor tyrosine kinase 3 Homo sapiens 217-243 21469976-2 2011 Sunitinib is an oral multitargeted inhibitor of vascular endothelial growth factor receptors (VEGFRs-1, -2, and -3), platelet-derived growth factor receptors (PDGFRs-alpha and -beta), stem-cell factor receptor (KIT), FMS-like tyrosine kinase 3 (FLT3), colony-stimulating factor 1 receptor (CSF-1R), and glial cell line-derived neurotrophic factor receptor (REarranged during Transfection; RET). Sunitinib 0-9 fms related receptor tyrosine kinase 3 Homo sapiens 245-249 21469976-2 2011 Sunitinib is an oral multitargeted inhibitor of vascular endothelial growth factor receptors (VEGFRs-1, -2, and -3), platelet-derived growth factor receptors (PDGFRs-alpha and -beta), stem-cell factor receptor (KIT), FMS-like tyrosine kinase 3 (FLT3), colony-stimulating factor 1 receptor (CSF-1R), and glial cell line-derived neurotrophic factor receptor (REarranged during Transfection; RET). Sunitinib 0-9 colony stimulating factor 1 receptor Homo sapiens 252-288 21469976-2 2011 Sunitinib is an oral multitargeted inhibitor of vascular endothelial growth factor receptors (VEGFRs-1, -2, and -3), platelet-derived growth factor receptors (PDGFRs-alpha and -beta), stem-cell factor receptor (KIT), FMS-like tyrosine kinase 3 (FLT3), colony-stimulating factor 1 receptor (CSF-1R), and glial cell line-derived neurotrophic factor receptor (REarranged during Transfection; RET). Sunitinib 0-9 colony stimulating factor 1 receptor Homo sapiens 290-296 21469976-2 2011 Sunitinib is an oral multitargeted inhibitor of vascular endothelial growth factor receptors (VEGFRs-1, -2, and -3), platelet-derived growth factor receptors (PDGFRs-alpha and -beta), stem-cell factor receptor (KIT), FMS-like tyrosine kinase 3 (FLT3), colony-stimulating factor 1 receptor (CSF-1R), and glial cell line-derived neurotrophic factor receptor (REarranged during Transfection; RET). Sunitinib 0-9 ret proto-oncogene Homo sapiens 389-392 21266595-0 2011 Sorafenib and sunitinib, two anticancer drugs, inhibit CYP3A4-mediated and activate CY3A5-mediated midazolam 1"-hydroxylation. Sunitinib 14-23 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 55-61 21266595-3 2011 The effects of sorafenib and sunitinib on midazolam 1"-hydroxylation catalyzed by human CYP3A4 or CYP3A5 were investigated. Sunitinib 29-38 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 88-94 21266595-4 2011 Sorafenib and sunitinib inhibited metabolic reactions catalyzed by recombinant CYP3A4. Sunitinib 14-23 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 79-85 21240876-13 2011 SUT significantly improved overexpression of dialysate transforming growth factor-ss1, monocyte chemoattractant protein-1 and vascular endothelial growth factor (VEGF) levels as compared with resting group. Sunitinib 0-3 C-C motif chemokine ligand 2 Rattus norvegicus 87-121 20809412-0 2011 Role of sorafenib and sunitinib in the induction of expressions of NKG2D ligands in nasopharyngeal carcinoma with high expression of ABCG2. Sunitinib 22-31 killer cell lectin like receptor K1 Homo sapiens 67-72 20809412-0 2011 Role of sorafenib and sunitinib in the induction of expressions of NKG2D ligands in nasopharyngeal carcinoma with high expression of ABCG2. Sunitinib 22-31 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 133-138 20809412-6 2011 FCM was used to evaluate the expressions of NKG2D ligands (NKG2DLs,) on target cells before and after incubated with sorafenib and sunitinib. Sunitinib 131-140 killer cell lectin like receptor K1 Homo sapiens 44-49 20809412-8 2011 RESULTS: The results revealed that target cells" cytotoxic sensitivity to natural killer (NK) cells increased in association with up-regulation of NKG2DLs on tumor cells after incubation with sorafenib and sunitinib. Sunitinib 206-215 killer cell lectin like receptor C1 Homo sapiens 147-151 20809412-9 2011 Furthermore, up-regulation in sunitinib group was much higher than in sorafenib group when it came to the expressions of NKG2DLs on tumor cells. Sunitinib 30-39 killer cell lectin like receptor C1 Homo sapiens 121-125 20809412-11 2011 CONCLUSIONS: Our research revealed for the first time that sorafenib and sunitinib could up-regulate NKG2DLs on tumor cells resulting in markedly increased tumor cells cytotoxic sensitivity to NK cells, which suggested that combining usage of molecular targeted agents and ACI may result in great benefits in clinical practice for the therapy-resistant cases and drug-resistant relapse. Sunitinib 73-82 killer cell lectin like receptor C1 Homo sapiens 101-105 21240876-13 2011 SUT significantly improved overexpression of dialysate transforming growth factor-ss1, monocyte chemoattractant protein-1 and vascular endothelial growth factor (VEGF) levels as compared with resting group. Sunitinib 0-3 vascular endothelial growth factor A Rattus norvegicus 126-160 21240876-13 2011 SUT significantly improved overexpression of dialysate transforming growth factor-ss1, monocyte chemoattractant protein-1 and vascular endothelial growth factor (VEGF) levels as compared with resting group. Sunitinib 0-3 vascular endothelial growth factor A Rattus norvegicus 162-166 21463120-2 2011 We conducted a phase II trial of the multi-targeted vascular endothelial growth factor receptor (VEGFR) kinase inhibitor, sunitinib, 37.5 mg given orally once daily in adult patients with relapsed or refractory DLBCL. Sunitinib 122-131 kinase insert domain receptor Homo sapiens 52-95 21463120-2 2011 We conducted a phase II trial of the multi-targeted vascular endothelial growth factor receptor (VEGFR) kinase inhibitor, sunitinib, 37.5 mg given orally once daily in adult patients with relapsed or refractory DLBCL. Sunitinib 122-131 kinase insert domain receptor Homo sapiens 97-102 20943595-12 2011 A concurrent fall in HPCs and growth factor expression (IGF-1) with sunitinib occurs. Sunitinib 68-77 insulin like growth factor 1 Homo sapiens 56-61 21343371-0 2011 IL6-STAT3-HIF signaling and therapeutic response to the angiogenesis inhibitor sunitinib in ovarian clear cell cancer. Sunitinib 79-88 interleukin 6 Homo sapiens 0-3 21343371-0 2011 IL6-STAT3-HIF signaling and therapeutic response to the angiogenesis inhibitor sunitinib in ovarian clear cell cancer. Sunitinib 79-88 signal transducer and activator of transcription 3 Homo sapiens 4-9 21309545-8 2011 Furthermore, sunitinib was transported equally by ABCB1 and ABCG2 at low concentrations, but ABCG2-mediated transport became saturated at lower concentrations than ABCB1-mediated transport. Sunitinib 13-22 ATP-binding cassette, sub-family B (MDR/TAP), member 1B Mus musculus 50-55 21309545-8 2011 Furthermore, sunitinib was transported equally by ABCB1 and ABCG2 at low concentrations, but ABCG2-mediated transport became saturated at lower concentrations than ABCB1-mediated transport. Sunitinib 13-22 ATP binding cassette subfamily G member 2 (Junior blood group) Mus musculus 60-65 20943595-2 2011 We investigate their expression in mRCC patients treated with sunitinib and correlate their expression with plasma growth factor levels [insulin-like growth factor (IGF)-1]. Sunitinib 62-71 insulin like growth factor 1 Homo sapiens 137-171 21325074-2 2011 Sunitinib, a potent inhibitor of RET, VEGF receptor (VEGFR)-1, VEGFR-2, VEGFR-3, and platelet-derived growth factor receptor (PDGFR)alpha/beta, has been reported as clinically effective in some patients with advanced MTC. Sunitinib 0-9 ret proto-oncogene Homo sapiens 33-36 20943595-8 2011 Both HPC and IGF-1 levels fell with sunitinib (61% and 14% fall, respectively, P <0.05 for both). Sunitinib 36-45 insulin like growth factor 1 Homo sapiens 13-18 21508369-11 2011 The amounts of p-VEGFR-2 and p-Tyr, as determined by immunohistochemistry, were greatly reduced in sunitinib-treated mice. Sunitinib 99-108 kinase insert domain protein receptor Mus musculus 17-24 21508369-12 2011 CONCLUSION: In a mouse model of mammary cancer, sunitinib inhibited tumor growth and metastasis (with the exception of lymph node metastasis), angiogenesis, and cell proliferation possibly due to reduced levels of p-VEGFR-2 and p-Tyr. Sunitinib 48-57 kinase insert domain protein receptor Mus musculus 216-223 21447721-2 2011 Sorafenib and sunitinib are small molecule inhibitors of multiple kinases including VEGF receptor (VEGFR) kinases. Sunitinib 14-23 kinase insert domain receptor Homo sapiens 84-97 21325074-2 2011 Sunitinib, a potent inhibitor of RET, VEGF receptor (VEGFR)-1, VEGFR-2, VEGFR-3, and platelet-derived growth factor receptor (PDGFR)alpha/beta, has been reported as clinically effective in some patients with advanced MTC. Sunitinib 0-9 fms related receptor tyrosine kinase 1 Homo sapiens 53-61 21447721-2 2011 Sorafenib and sunitinib are small molecule inhibitors of multiple kinases including VEGF receptor (VEGFR) kinases. Sunitinib 14-23 kinase insert domain receptor Homo sapiens 99-104 21325074-2 2011 Sunitinib, a potent inhibitor of RET, VEGF receptor (VEGFR)-1, VEGFR-2, VEGFR-3, and platelet-derived growth factor receptor (PDGFR)alpha/beta, has been reported as clinically effective in some patients with advanced MTC. Sunitinib 0-9 kinase insert domain receptor Homo sapiens 63-70 21325074-2 2011 Sunitinib, a potent inhibitor of RET, VEGF receptor (VEGFR)-1, VEGFR-2, VEGFR-3, and platelet-derived growth factor receptor (PDGFR)alpha/beta, has been reported as clinically effective in some patients with advanced MTC. Sunitinib 0-9 fms related receptor tyrosine kinase 4 Homo sapiens 72-79 21325074-2 2011 Sunitinib, a potent inhibitor of RET, VEGF receptor (VEGFR)-1, VEGFR-2, VEGFR-3, and platelet-derived growth factor receptor (PDGFR)alpha/beta, has been reported as clinically effective in some patients with advanced MTC. Sunitinib 0-9 platelet derived growth factor receptor alpha Homo sapiens 126-137 21325074-4 2011 EXPERIMENTAL DESIGN: Both in vitro and in vivo assays, using the human TT RET(C634W) MTC cell line, were done to assess the activity of sunitinib. Sunitinib 136-145 ret proto-oncogene Homo sapiens 74-77 21325074-6 2011 RESULTS: Sunitinib displayed antiproliferative and antiangiogenic activities and inhibited RET autophosphorylation and activation of downstream signaling pathways. Sunitinib 9-18 ret proto-oncogene Homo sapiens 91-94 21325074-7 2011 We showed that sunitinib treatment induced major changes in the expression of genes involved in tissue invasion and metastasis including vimentin (VIM), urokinase plasminogen (PLAU), tenascin-C (TN-C), SPARC, and CD44. Sunitinib 15-24 vimentin Homo sapiens 137-145 21325074-7 2011 We showed that sunitinib treatment induced major changes in the expression of genes involved in tissue invasion and metastasis including vimentin (VIM), urokinase plasminogen (PLAU), tenascin-C (TN-C), SPARC, and CD44. Sunitinib 15-24 vimentin Homo sapiens 147-150 21325074-7 2011 We showed that sunitinib treatment induced major changes in the expression of genes involved in tissue invasion and metastasis including vimentin (VIM), urokinase plasminogen (PLAU), tenascin-C (TN-C), SPARC, and CD44. Sunitinib 15-24 plasminogen activator, urokinase Homo sapiens 176-180 21325074-7 2011 We showed that sunitinib treatment induced major changes in the expression of genes involved in tissue invasion and metastasis including vimentin (VIM), urokinase plasminogen (PLAU), tenascin-C (TN-C), SPARC, and CD44. Sunitinib 15-24 tenascin C Homo sapiens 183-193 21325074-7 2011 We showed that sunitinib treatment induced major changes in the expression of genes involved in tissue invasion and metastasis including vimentin (VIM), urokinase plasminogen (PLAU), tenascin-C (TN-C), SPARC, and CD44. Sunitinib 15-24 tenascin C Homo sapiens 195-199 21325074-7 2011 We showed that sunitinib treatment induced major changes in the expression of genes involved in tissue invasion and metastasis including vimentin (VIM), urokinase plasminogen (PLAU), tenascin-C (TN-C), SPARC, and CD44. Sunitinib 15-24 secreted protein acidic and cysteine rich Homo sapiens 202-207 21325074-7 2011 We showed that sunitinib treatment induced major changes in the expression of genes involved in tissue invasion and metastasis including vimentin (VIM), urokinase plasminogen (PLAU), tenascin-C (TN-C), SPARC, and CD44. Sunitinib 15-24 CD44 molecule (Indian blood group) Homo sapiens 213-217 21325074-8 2011 Analyzing downregulated genes, we identified those encoding secreted proteins and, among them, interleukin (IL)-8 was found to be modulated in the serum of xenografted mice under sunitinib treatment. Sunitinib 179-188 chemokine (C-X-C motif) ligand 15 Mus musculus 95-113 21540050-8 2011 Additional broad-spectrum VEGF receptor tyrosine kinase inhibitors, such as sunitinib and sorafenib, are used in monotherapy for metastatic renal carcinoma, while sunitinib is also approved for imatinib resistant gastrointestinal stromal tumors and sorafenib for advanced stage hepatocellular carcinoma. Sunitinib 76-85 vascular endothelial growth factor A Homo sapiens 26-30 21220434-5 2011 The sunitinib PK study was designed to determine the relationship between CYP3A4 activity and sunitinib exposure using 7.5 mg midazolam orally as a phenotypic probe. Sunitinib 4-13 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 74-80 21220434-5 2011 The sunitinib PK study was designed to determine the relationship between CYP3A4 activity and sunitinib exposure using 7.5 mg midazolam orally as a phenotypic probe. Sunitinib 94-103 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 74-80 21097529-5 2011 We further present evidence that L1-CAM was involved in the resistance against therapeutic reagents like rapamycin, sunitinib and cisplatin. Sunitinib 116-125 L1 cell adhesion molecule Homo sapiens 33-39 21057538-0 2011 Fibroblast growth factor 2 regulates endothelial cell sensitivity to sunitinib. Sunitinib 69-78 fibroblast growth factor 2 Homo sapiens 0-26 21057538-1 2011 The vascular endothelial growth factor (VEGF) receptor tyrosine kinase inhibitor sunitinib has been approved for first-line treatment of patients with metastatic renal cancer and is currently being trialled in other cancers. Sunitinib 81-90 vascular endothelial growth factor A Homo sapiens 4-38 21057538-1 2011 The vascular endothelial growth factor (VEGF) receptor tyrosine kinase inhibitor sunitinib has been approved for first-line treatment of patients with metastatic renal cancer and is currently being trialled in other cancers. Sunitinib 81-90 vascular endothelial growth factor A Homo sapiens 40-44 21057538-3 2011 By screening a panel of candidate growth factors we identified fibroblast growth factor 2 (FGF2) as a potent regulator of endothelial cell sensitivity to sunitinib. Sunitinib 154-163 fibroblast growth factor 2 Homo sapiens 63-89 21057538-3 2011 By screening a panel of candidate growth factors we identified fibroblast growth factor 2 (FGF2) as a potent regulator of endothelial cell sensitivity to sunitinib. Sunitinib 154-163 fibroblast growth factor 2 Homo sapiens 91-95 21057538-4 2011 We show that FGF2 supports endothelial proliferation and de novo tubule formation in the presence of sunitinib and that FGF2 can suppress sunitinib-induced retraction of tubules. Sunitinib 101-110 fibroblast growth factor 2 Homo sapiens 13-17 21057538-4 2011 We show that FGF2 supports endothelial proliferation and de novo tubule formation in the presence of sunitinib and that FGF2 can suppress sunitinib-induced retraction of tubules. Sunitinib 138-147 fibroblast growth factor 2 Homo sapiens 120-124 21317875-4 2011 We show that the Xbp1-specific ribonuclease activity depends on autophosphorylation, and that ATP-competitive inhibitors staurosporin and sunitinib, which inhibit autophosphorylation in vitro, also inhibit Xbp1 splicing in vivo. Sunitinib 138-147 X-box binding protein 1 Homo sapiens 17-21 21317875-4 2011 We show that the Xbp1-specific ribonuclease activity depends on autophosphorylation, and that ATP-competitive inhibitors staurosporin and sunitinib, which inhibit autophosphorylation in vitro, also inhibit Xbp1 splicing in vivo. Sunitinib 138-147 X-box binding protein 1 Homo sapiens 206-210 20437403-5 2011 The activation of this pathway explains the use of both VEGF and VEGF-receptor inhibitors (bevacizumab, sunitinib and sorafenib) in the therapy of advanced CCRCC. Sunitinib 104-113 vascular endothelial growth factor A Homo sapiens 65-69 21308478-2 2011 Recently, data from a phase III trial demonstrated that sunitinib produces a clinically significant improvement in progression-free survival in patients with unresectable, advanced, or metastatic GEP-NETs. Sunitinib 56-65 granulin precursor Homo sapiens 196-199 21308478-3 2011 Based on this finding, sunitinib became the first targeted drug approved for the treatment of GEP-NETs, paving the way for the approval of other anticancer agents in this drug-orphan disease. Sunitinib 23-32 granulin precursor Homo sapiens 94-97 21233403-10 2011 When combined with sunitinib, sorafenib, bevacizumab, irinotecan, or docetaxel, Ang2 CovX-Bodies produced even greater efficacy (~80% TGI, P < 0.01). Sunitinib 19-28 angiopoietin 2 Homo sapiens 80-84 20437403-5 2011 The activation of this pathway explains the use of both VEGF and VEGF-receptor inhibitors (bevacizumab, sunitinib and sorafenib) in the therapy of advanced CCRCC. Sunitinib 104-113 vascular endothelial growth factor A Homo sapiens 56-60 21097719-3 2011 PDGFRbeta was found to be the main tyrosine kinase target of sunitinib expressed in BM stromal ST-2 and MC3T3-E1 preosteoblastic cells. Sunitinib 61-70 platelet derived growth factor receptor, beta polypeptide Mus musculus 0-9 20708948-3 2011 Despite the initial unimpressive clinical performance of anti-VEGF antibody (bevacizumab) as cancer monotherapy, clear improvements in clinical outcomes following combination bevacizumab and chemotherapy regimens and multi-targeted VEGF receptor tyrosine kinase inhibitors (sorafenib and sunitinib) in select tumor types have established VEGF-targeted agents as an effective means of controlling cancer growth. Sunitinib 288-297 vascular endothelial growth factor A Homo sapiens 232-236 20708948-3 2011 Despite the initial unimpressive clinical performance of anti-VEGF antibody (bevacizumab) as cancer monotherapy, clear improvements in clinical outcomes following combination bevacizumab and chemotherapy regimens and multi-targeted VEGF receptor tyrosine kinase inhibitors (sorafenib and sunitinib) in select tumor types have established VEGF-targeted agents as an effective means of controlling cancer growth. Sunitinib 288-297 vascular endothelial growth factor A Homo sapiens 232-236 21273619-2 2011 Limited clinical trial data have shown minimal activity with cytokines and chemotherapy, although small-molecule inhibitors of the vascular endothelial growth factor and platelet-derived growth factor pathways such as sunitinib and sorafenib, which are associated with significant clinical activity in clear-cell RCC (ccRCC), have been associated with a 25% response rate in chRCC. Sunitinib 218-227 vascular endothelial growth factor A Homo sapiens 131-165 21084271-0 2011 VEGF-PET imaging is a noninvasive biomarker showing differential changes in the tumor during sunitinib treatment. Sunitinib 93-102 vascular endothelial growth factor A Homo sapiens 0-4 21084271-2 2011 In this study, we evaluated the role of VEGF-PET as a biomarker of dynamic angiogenic changes in tumors following treatment with the kinase inhibitor sunitinib. Sunitinib 150-159 vascular endothelial growth factor A Homo sapiens 40-44 21484496-2 2011 The first category, vascular endothelial growth factor (VEGF)-directed therapies, includes sunitinib, pazopanib, sorafenib and bevacizumab. Sunitinib 91-100 vascular endothelial growth factor A Homo sapiens 20-54 21484496-2 2011 The first category, vascular endothelial growth factor (VEGF)-directed therapies, includes sunitinib, pazopanib, sorafenib and bevacizumab. Sunitinib 91-100 vascular endothelial growth factor A Homo sapiens 56-60 22130231-14 2011 VEGFR tyrosine kinase inhibitors such as sunitinib and sorafenib are also effective in antiangiogenic tumor therapy by inhibiting VEGFR signaling. Sunitinib 41-50 kinase insert domain receptor Homo sapiens 0-5 22130231-14 2011 VEGFR tyrosine kinase inhibitors such as sunitinib and sorafenib are also effective in antiangiogenic tumor therapy by inhibiting VEGFR signaling. Sunitinib 41-50 kinase insert domain receptor Homo sapiens 130-135 25819130-3 2011 For sunitinib, sorafenib, and pazopanib: potential drug interactions are possible with inducers/inhibitors of CYP3A4, anti-hypertensive drugs, antidiabetic drugs, thyroid hormones, and anticoagulant treatments. Sunitinib 4-13 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 110-116 21116624-15 2011 The mechanism of resistance to sunitinib is unknown at this time but is also appears related to growth of clones with secondary mutations in KIT. Sunitinib 31-40 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 141-144 21084271-5 2011 In contrast to (18)F-FDG and (15)O-water PET, VEGF-PET demonstrated dynamic changes during sunitinib treatment within the tumor with a strong decline in signal in the tumor center and only minimal reduction in tumor rim, with a pronounced rebound after sunitinib discontinuation. Sunitinib 91-100 vascular endothelial growth factor A Homo sapiens 46-50 21097719-3 2011 PDGFRbeta was found to be the main tyrosine kinase target of sunitinib expressed in BM stromal ST-2 and MC3T3-E1 preosteoblastic cells. Sunitinib 61-70 interleukin 1 receptor-like 1 Mus musculus 95-99 21097719-5 2011 Incubation of ST-2 and human BM endothelial cells with sunitinib led to potent cell growth inhibition and induction of apoptosis in a dose-dependent manner. Sunitinib 55-64 ST2 Homo sapiens 14-18 21181476-7 2011 Sunitinib malate, a second-generation KIT inhibitor is active in imatinib-resistant disease and is FDA-approved for use in this setting. Sunitinib 0-16 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 38-41 21097719-6 2011 Similarly, sunitinib induced a robust proapoptotic effect in vivo on BM stromal PDGFRbeta(+) cells and produced extensive disruption of tissue architecture and vessel leakage in the BM cavity. Sunitinib 11-20 platelet derived growth factor receptor, beta polypeptide Mus musculus 80-89 21097719-7 2011 Pretreatment of ST-2 cells with sunitinib also hindered heterotypic adhesion to lung cancer cell lines. Sunitinib 32-41 interleukin 1 receptor-like 1 Mus musculus 16-20 21627339-2 2011 Immunotherapy with interferon-alpha (IFNalpha) and interleukin-2 (IL-2) has been the historical therapy of choice for the treatment of locally advanced or metastatic RCC prior to the more recent development of targeted therapies, including sunitinib and bevacizumab (combined with IFNalpha). Sunitinib 240-249 interleukin 2 Homo sapiens 66-70 22050750-9 2011 Other small molecule inhibitors of VEGF tyrosine kinase activity (TKIs) such as sunitinib, vandetanib and sorafenib are being tested in MBC. Sunitinib 80-89 vascular endothelial growth factor A Homo sapiens 35-39 21717054-2 2011 Sunitinib is able to inhibit RTKs such as receptors for platelet-derived growth factor (PDGF-Ralpha and beta) and for vascular endothelial growth factor (VEGFRs). Sunitinib 0-9 vascular endothelial growth factor A Homo sapiens 118-152 20304669-7 2011 Since c-kit can be readily inhibited by several small molecule inhibitors including imatinib, sunitinib and dasatinib, targeting immune suppressing cells can be readily accomplished in the clinic. Sunitinib 94-103 KIT proto-oncogene receptor tyrosine kinase Mus musculus 6-11 20524040-0 2011 Sunitinib induces PTEN expression and inhibits PDGFR signaling and migration of medulloblastoma cells. Sunitinib 0-9 phosphatase and tensin homolog Homo sapiens 18-22 20524040-0 2011 Sunitinib induces PTEN expression and inhibits PDGFR signaling and migration of medulloblastoma cells. Sunitinib 0-9 platelet derived growth factor receptor beta Homo sapiens 47-52 20524040-3 2011 In this study, we investigated whether the multi-tyrosine kinase inhibitor sunitinib, effectively inhibits PDGFR signaling required for medulloblastoma cell migration. Sunitinib 75-84 platelet derived growth factor receptor beta Homo sapiens 107-112 20524040-4 2011 Daoy and D556 human medulloblastoma cells pre-treated for 1 h with 0.2 muM sunitinib demonstrated induction of PTEN expression and significant inhibition of PDGFR signaling activity and transactivation of EGFR, in a RAS-independent manner, in response to PDGF-BB stimulation. Sunitinib 75-84 phosphatase and tensin homolog Homo sapiens 111-115 20524040-4 2011 Daoy and D556 human medulloblastoma cells pre-treated for 1 h with 0.2 muM sunitinib demonstrated induction of PTEN expression and significant inhibition of PDGFR signaling activity and transactivation of EGFR, in a RAS-independent manner, in response to PDGF-BB stimulation. Sunitinib 75-84 platelet derived growth factor receptor beta Homo sapiens 157-162 20524040-4 2011 Daoy and D556 human medulloblastoma cells pre-treated for 1 h with 0.2 muM sunitinib demonstrated induction of PTEN expression and significant inhibition of PDGFR signaling activity and transactivation of EGFR, in a RAS-independent manner, in response to PDGF-BB stimulation. Sunitinib 75-84 epidermal growth factor receptor Homo sapiens 205-209 20524040-8 2011 Sunitinib similarly inhibited PDGFR signaling and migration of primary murine Smo/Smo medulloblastoma cells, suggesting that the Smo/Smo mouse is an appropriate model for preclinical testing of sunitinib. Sunitinib 0-9 platelet derived growth factor receptor, beta polypeptide Mus musculus 30-35 20524040-8 2011 Sunitinib similarly inhibited PDGFR signaling and migration of primary murine Smo/Smo medulloblastoma cells, suggesting that the Smo/Smo mouse is an appropriate model for preclinical testing of sunitinib. Sunitinib 0-9 smoothened, frizzled class receptor Mus musculus 78-81 20524040-8 2011 Sunitinib similarly inhibited PDGFR signaling and migration of primary murine Smo/Smo medulloblastoma cells, suggesting that the Smo/Smo mouse is an appropriate model for preclinical testing of sunitinib. Sunitinib 0-9 smoothened, frizzled class receptor Mus musculus 82-85 20524040-8 2011 Sunitinib similarly inhibited PDGFR signaling and migration of primary murine Smo/Smo medulloblastoma cells, suggesting that the Smo/Smo mouse is an appropriate model for preclinical testing of sunitinib. Sunitinib 0-9 smoothened, frizzled class receptor Mus musculus 82-85 20524040-8 2011 Sunitinib similarly inhibited PDGFR signaling and migration of primary murine Smo/Smo medulloblastoma cells, suggesting that the Smo/Smo mouse is an appropriate model for preclinical testing of sunitinib. Sunitinib 0-9 smoothened, frizzled class receptor Mus musculus 82-85 21187670-10 2011 The activity of PI3K/Akt signal pathway was inhibited, which could be the potential mechanisms that account for the enhanced radiation response induced by sunitinib. Sunitinib 155-164 AKT serine/threonine kinase 1 Homo sapiens 21-24 21980525-10 2011 In agreement, we observed that the binding of VEGFR2 to DNA requires VEGF activation, being blocked by Bevacizumab and Sunitinib, two anti-angiogenic agents that inhibit VEGFR2 activation. Sunitinib 119-128 kinase insert domain receptor Homo sapiens 46-52 21344032-9 2011 Sunitinib suppressed the expression of cyclin B1, p-Akt, and t-Akt while augmenting the expression of cyclin D and pro-apoptotic Bax and Bad in HTB5 cells. Sunitinib 0-9 cyclin B1 Homo sapiens 39-48 21344032-9 2011 Sunitinib suppressed the expression of cyclin B1, p-Akt, and t-Akt while augmenting the expression of cyclin D and pro-apoptotic Bax and Bad in HTB5 cells. Sunitinib 0-9 AKT serine/threonine kinase 1 Homo sapiens 52-55 21344032-9 2011 Sunitinib suppressed the expression of cyclin B1, p-Akt, and t-Akt while augmenting the expression of cyclin D and pro-apoptotic Bax and Bad in HTB5 cells. Sunitinib 0-9 AKT serine/threonine kinase 1 Homo sapiens 63-66 21344032-9 2011 Sunitinib suppressed the expression of cyclin B1, p-Akt, and t-Akt while augmenting the expression of cyclin D and pro-apoptotic Bax and Bad in HTB5 cells. Sunitinib 0-9 BCL2 associated X, apoptosis regulator Homo sapiens 129-132 21625184-0 2011 Efficacy of sunitinib re-exposure after failure of an mTOR inhibitor in patients with metastatic RCC. Sunitinib 12-21 mechanistic target of rapamycin kinase Homo sapiens 54-58 21245940-5 2011 Sunitinib (40 mg/kg, once daily), a receptor tyrosine kinase inhibitor of VEGFR2 and other growth factor receptors, also caused significant loss of tumor blood vessels in RCC models but had weaker effects than DC101 on pericytes and lymphatic vessels. Sunitinib 0-9 kinase insert domain protein receptor Mus musculus 74-80 21980525-10 2011 In agreement, we observed that the binding of VEGFR2 to DNA requires VEGF activation, being blocked by Bevacizumab and Sunitinib, two anti-angiogenic agents that inhibit VEGFR2 activation. Sunitinib 119-128 vascular endothelial growth factor A Homo sapiens 46-50 21980525-10 2011 In agreement, we observed that the binding of VEGFR2 to DNA requires VEGF activation, being blocked by Bevacizumab and Sunitinib, two anti-angiogenic agents that inhibit VEGFR2 activation. Sunitinib 119-128 kinase insert domain receptor Homo sapiens 170-176 20975250-7 2011 RESULTS: Sunitinib was associated with significant declines in total leucocyte (-48%), neutrophil (-62%), CD3 total T cell (-31%) and CD4 counts (32%; p < 0.05). Sunitinib 9-18 CD4 molecule Homo sapiens 134-137 20818610-2 2010 Small molecule kinase inhibitors have been developed for the treatment of certain cancers, and some antioncogenic agents such as sunitinib, may nonspecifically inhibit LRRK2. Sunitinib 129-138 leucine rich repeat kinase 2 Homo sapiens 168-173 21187479-1 2010 OBJECTIVE: The aim of this study was to assess the antitumour effect of an anti-VEGFR (sunitinib) and the anti-EGFR multi-targeted agent (lapatinib), applied either alone or in combination on the migration capacity of two glioma cell lines. Sunitinib 87-96 kinase insert domain receptor Homo sapiens 80-85 21187479-1 2010 OBJECTIVE: The aim of this study was to assess the antitumour effect of an anti-VEGFR (sunitinib) and the anti-EGFR multi-targeted agent (lapatinib), applied either alone or in combination on the migration capacity of two glioma cell lines. Sunitinib 87-96 epidermal growth factor receptor Homo sapiens 81-85 21187479-2 2010 Furthermore, this study sought to evaluate the effect of lapatinib in the formation of EGFR-integrin beta(1) complex, as well as the effect of sunitinib in the VEGFR-integrin beta(3) and PDGFR-integrin beta(3) complexes formation. Sunitinib 143-152 kinase insert domain receptor Homo sapiens 160-165 21187479-9 2010 Likewise, sunitinib inhibited complex formation of VEGFR-integrin beta(3) complex within two h after its application without affecting PDGFR-integrin beta(3) complex. Sunitinib 10-19 kinase insert domain receptor Homo sapiens 51-56 21187479-11 2010 CONCLUSION: The preliminary results of this study are the first to support the implication of a dual anti-EGFR/HER-2 agent, lapatinib and a multi-targeted agent, sunitinib in glioma cell migration, through a mechanism implying interruption of growth factor receptor integrin complexes formation. Sunitinib 162-171 epidermal growth factor receptor Homo sapiens 106-110 21118964-6 2010 Sunitinib inhibited Stat3 in dendritic cells and T cells and reduced conversion of transferred FoxP3(-) T cells to tumor-associated regulatory T cells while increasing transferred CD8(+) T-cell infiltration and activation at the tumor site, leading to inhibition of primary tumor growth. Sunitinib 0-9 signal transducer and activator of transcription 3 Homo sapiens 20-25 21118964-6 2010 Sunitinib inhibited Stat3 in dendritic cells and T cells and reduced conversion of transferred FoxP3(-) T cells to tumor-associated regulatory T cells while increasing transferred CD8(+) T-cell infiltration and activation at the tumor site, leading to inhibition of primary tumor growth. Sunitinib 0-9 forkhead box P3 Homo sapiens 95-100 21118964-6 2010 Sunitinib inhibited Stat3 in dendritic cells and T cells and reduced conversion of transferred FoxP3(-) T cells to tumor-associated regulatory T cells while increasing transferred CD8(+) T-cell infiltration and activation at the tumor site, leading to inhibition of primary tumor growth. Sunitinib 0-9 CD8a molecule Homo sapiens 180-183 20952508-0 2010 HGF/c-Met acts as an alternative angiogenic pathway in sunitinib-resistant tumors. Sunitinib 55-64 hepatocyte growth factor Homo sapiens 0-3 20952508-0 2010 HGF/c-Met acts as an alternative angiogenic pathway in sunitinib-resistant tumors. Sunitinib 55-64 MET proto-oncogene, receptor tyrosine kinase Homo sapiens 4-9 20952508-6 2010 Combination of sunitinib and a selective c-Met inhibitor significantly inhibited tumor growth compared with sunitinib or c-Met inhibitor alone in resistant tumors. Sunitinib 108-117 MET proto-oncogene, receptor tyrosine kinase Homo sapiens 41-46 20952508-8 2010 Conversely, systemic injection of HGF in the sensitive tumor models conferred resistance to sunitinib through maintenance of tumor angiogenesis. Sunitinib 92-101 hepatocyte growth factor Homo sapiens 34-37 20142724-1 2010 OBJECTIVES: Sunitinib is an oral, multitargeted tyrosine kinase inhibitor of vascular endothelial growth factor and platelet-derived growth factor receptors with proven clinical benefit in patients with metastatic renal cell carcinoma (RCC). Sunitinib 12-21 vascular endothelial growth factor A Homo sapiens 77-111 21095547-4 2010 Recently, sunitinib demonstrating efficacy in pancreatic islet cell carcinomas has opened a new avenue for the treatment of NETs, and further trials shall be considered in NET types such as carcinoids, poorly differentiated neuroendocrine carcinomas, and several other endocrine tumors that depend on vascular endothelial growth factor (VEGF)/VEGF receptor for angiogenesis. Sunitinib 10-19 vascular endothelial growth factor A Homo sapiens 301-335 21095547-4 2010 Recently, sunitinib demonstrating efficacy in pancreatic islet cell carcinomas has opened a new avenue for the treatment of NETs, and further trials shall be considered in NET types such as carcinoids, poorly differentiated neuroendocrine carcinomas, and several other endocrine tumors that depend on vascular endothelial growth factor (VEGF)/VEGF receptor for angiogenesis. Sunitinib 10-19 vascular endothelial growth factor A Homo sapiens 337-341 21095547-4 2010 Recently, sunitinib demonstrating efficacy in pancreatic islet cell carcinomas has opened a new avenue for the treatment of NETs, and further trials shall be considered in NET types such as carcinoids, poorly differentiated neuroendocrine carcinomas, and several other endocrine tumors that depend on vascular endothelial growth factor (VEGF)/VEGF receptor for angiogenesis. Sunitinib 10-19 vascular endothelial growth factor A Homo sapiens 343-347 21030045-0 2010 The immunocytokine F8-IL2 improves the therapeutic performance of sunitinib in a mouse model of renal cell carcinoma. Sunitinib 66-75 interleukin-2 Cricetulus griseus 22-25 21030045-6 2010 CONCLUSIONS: Considering that recombinant interleukin-2 based immunocytokines are now being investigated in several clinical trials in patients with cancer alone or combined with chemotherapy our preclinical results provide a motivation to study F8-IL2 combined with sunitinib in clinical trials in patients with kidney cancer. Sunitinib 267-276 interleukin 2 Homo sapiens 42-55 20948437-5 2010 We observed a decrease in the number of peripheral blood Foxp3+ regulatory T cells after each cycle of sunitinib-based therapy. Sunitinib 103-112 forkhead box P3 Homo sapiens 57-62 20950441-7 2010 After paracetamol coadministration with low sunitinib doses (group-D), ALT and AST concentrations ranged 182.79-221.03 U/L and 259.7-264.4 U/L respectively, lower than group-B. Sunitinib 44-53 glutamic pyruvic transaminase, soluble Mus musculus 71-74 20471160-0 2010 mTOR inhibition by everolimus counteracts VEGF induction by sunitinib and improves anti-tumor activity against gastric cancer in vivo. Sunitinib 60-69 mechanistic target of rapamycin kinase Homo sapiens 0-4 20471160-0 2010 mTOR inhibition by everolimus counteracts VEGF induction by sunitinib and improves anti-tumor activity against gastric cancer in vivo. Sunitinib 60-69 vascular endothelial growth factor A Homo sapiens 42-46 20471160-2 2010 We hypothesized that mTOR inhibition by everolimus counteracts VEGF induction by sunitinib resulting in an improved anti-tumor activity of sunitinib. Sunitinib 81-90 mechanistic target of rapamycin kinase Homo sapiens 21-25 20471160-2 2010 We hypothesized that mTOR inhibition by everolimus counteracts VEGF induction by sunitinib resulting in an improved anti-tumor activity of sunitinib. Sunitinib 81-90 vascular endothelial growth factor A Homo sapiens 63-67 20471160-2 2010 We hypothesized that mTOR inhibition by everolimus counteracts VEGF induction by sunitinib resulting in an improved anti-tumor activity of sunitinib. Sunitinib 139-148 mechanistic target of rapamycin kinase Homo sapiens 21-25 20471160-2 2010 We hypothesized that mTOR inhibition by everolimus counteracts VEGF induction by sunitinib resulting in an improved anti-tumor activity of sunitinib. Sunitinib 139-148 vascular endothelial growth factor A Homo sapiens 63-67 20471160-6 2010 In conclusion mTOR inhibition by everolimus counteracts VEGF induction by sunitinib and results in significant reduction of tumor burden and long-lasting tumor growth control. Sunitinib 74-83 mechanistic target of rapamycin kinase Homo sapiens 14-18 20471160-6 2010 In conclusion mTOR inhibition by everolimus counteracts VEGF induction by sunitinib and results in significant reduction of tumor burden and long-lasting tumor growth control. Sunitinib 74-83 vascular endothelial growth factor A Homo sapiens 56-60 20950441-8 2010 Paracetamol coadministration with high sunitinib doses showed higher ALT and AST values (range 269.6-349.2 U/L and 430.2-540.3 U/L respectively), p < 0.05. Sunitinib 39-48 glutamic pyruvic transaminase, soluble Mus musculus 69-72 20950441-8 2010 Paracetamol coadministration with high sunitinib doses showed higher ALT and AST values (range 269.6-349.2 U/L and 430.2-540.3 U/L respectively), p < 0.05. Sunitinib 39-48 solute carrier family 17 (anion/sugar transporter), member 5 Mus musculus 77-80 20686367-1 2010 SU11248 is a selective inhibitor of certain protein kinases including VEGFR types 1-3 that are expressed in human breast cancer. Sunitinib 0-7 kinase insert domain receptor Homo sapiens 70-75 20591397-16 2010 Blocking both VEGF and platelet-derived growth factor pathways using sunitinib was 3-fold more effective. Sunitinib 69-78 vascular endothelial growth factor A Oryctolagus cuniculus 14-18 20943646-9 2010 The vascular endothelial growth factor inhibitor sunitinib has demonstrated antitumor effects in pancreatic NETs. Sunitinib 49-58 vascular endothelial growth factor A Homo sapiens 4-38 20686367-10 2010 VEGF (10 ng/ml) caused a 42% increase in proliferation of E0771 cells, compared to the control (p < 0.01; n = 8), and there was a significant decrease in proliferation of E0771 cells treated with VEGF plus SU11248 (10 mumol/L) vs. the control (65%, p < 0.01). Sunitinib 209-216 vascular endothelial growth factor A Homo sapiens 0-4 20686367-11 2010 VEGF caused a 2-fold increase in the proliferation of HUVEC vs. the control (p < 0.01; n = 8), but its action was completely eradicated by SU11248. Sunitinib 142-149 vascular endothelial growth factor A Homo sapiens 0-4 20686367-15 2010 These findings support the hypothesis that the antitumor activity of SU11248 on breast cancer is possibly mediated by targeting the paracrine and autocrine effects of VEGF on breast cancer to suppress tumor angiogenesis, proliferation and migration. Sunitinib 69-76 vascular endothelial growth factor A Homo sapiens 167-171 20133148-3 2010 Sunitinib is an oral multitargeted inhibitor of the VEGF, platelet-derived growth factor (PDGF), and c-KIT, among others, tyrosine kinase receptors. Sunitinib 0-9 vascular endothelial growth factor A Homo sapiens 52-56 20133148-3 2010 Sunitinib is an oral multitargeted inhibitor of the VEGF, platelet-derived growth factor (PDGF), and c-KIT, among others, tyrosine kinase receptors. Sunitinib 0-9 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 101-106 21090521-3 2010 Sunitinib and sorafenib are small molecule tyrosine kinase inhibitors that target vascular endothelial growth factor receptor, platelet-derived growth factor receptor, C-Kit and others. Sunitinib 0-9 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 168-173 20733093-0 2010 Hypertension induced by the tyrosine kinase inhibitor sunitinib is associated with increased circulating endothelin-1 levels. Sunitinib 54-63 endothelin 1 Homo sapiens 105-117 20733093-7 2010 In rats, 8 days of sunitinib administration induced an 30-mm Hg rise in BP, an attenuation of the circadian BP rhythm, and a 3-fold rise in serum endothelin-1 and creatinine, of which all but the rise in creatinine reversed after sunitinib withdrawal. Sunitinib 19-28 endothelin 1 Rattus norvegicus 147-159 20733093-8 2010 Coronary microvascular function studies after 8 days of sunitinib administration showed decreased responses to bradykinin, angiotensin II, and sodium nitroprusside, all normalizing after sunitinib withdrawal. Sunitinib 56-65 kininogen 1 Homo sapiens 111-121 20733093-8 2010 Coronary microvascular function studies after 8 days of sunitinib administration showed decreased responses to bradykinin, angiotensin II, and sodium nitroprusside, all normalizing after sunitinib withdrawal. Sunitinib 56-65 angiotensinogen Homo sapiens 123-137 20733093-10 2010 In conclusion, sunitinib induces a reversible rise in BP in patients and in rats associated with activation of the endothelin-1 system, suppression of the renin-angiotensin system, and generalized microvascular dysfunction. Sunitinib 15-24 endothelin 1 Rattus norvegicus 115-127 20733093-10 2010 In conclusion, sunitinib induces a reversible rise in BP in patients and in rats associated with activation of the endothelin-1 system, suppression of the renin-angiotensin system, and generalized microvascular dysfunction. Sunitinib 15-24 renin Rattus norvegicus 155-160 19603143-11 2010 However, MMP-9 and MMP-2 levels were decreased 48 h after treatment of M059K cells with sunitinib either alone or in combination with lapatinib. Sunitinib 88-97 matrix metallopeptidase 9 Homo sapiens 9-14 19603143-11 2010 However, MMP-9 and MMP-2 levels were decreased 48 h after treatment of M059K cells with sunitinib either alone or in combination with lapatinib. Sunitinib 88-97 matrix metallopeptidase 2 Homo sapiens 19-24 20699227-6 2010 Sunitinib administration led to an early downmodulation of IFNgamma levels as well as reduction of IFNgamma receptor and downstream angiostatic chemokines CXCL9 to 11 within the tumor. Sunitinib 0-9 interferon gamma Homo sapiens 59-67 19649772-1 2010 BACKGROUND: Sunitinib is an orally administered multitargeted tyrosine kinase inhibitor of RET, VEGFR, PDGFR, and c-KIT. Sunitinib 12-21 ret proto-oncogene Homo sapiens 91-94 19649772-1 2010 BACKGROUND: Sunitinib is an orally administered multitargeted tyrosine kinase inhibitor of RET, VEGFR, PDGFR, and c-KIT. Sunitinib 12-21 kinase insert domain receptor Homo sapiens 96-101 19649772-1 2010 BACKGROUND: Sunitinib is an orally administered multitargeted tyrosine kinase inhibitor of RET, VEGFR, PDGFR, and c-KIT. Sunitinib 12-21 platelet derived growth factor receptor beta Homo sapiens 103-108 19649772-1 2010 BACKGROUND: Sunitinib is an orally administered multitargeted tyrosine kinase inhibitor of RET, VEGFR, PDGFR, and c-KIT. Sunitinib 12-21 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 114-119 19649772-22 2010 Sunitinib was well tolerated in PS 2 patients. Sunitinib 0-9 taste 2 receptor member 64 pseudogene Homo sapiens 32-36 20699227-6 2010 Sunitinib administration led to an early downmodulation of IFNgamma levels as well as reduction of IFNgamma receptor and downstream angiostatic chemokines CXCL9 to 11 within the tumor. Sunitinib 0-9 interferon gamma Homo sapiens 99-107 20699227-6 2010 Sunitinib administration led to an early downmodulation of IFNgamma levels as well as reduction of IFNgamma receptor and downstream angiostatic chemokines CXCL9 to 11 within the tumor. Sunitinib 0-9 C-X-C motif chemokine ligand 9 Homo sapiens 155-160 20699227-7 2010 Intratumoral injection of CXCL9, although producing minimal effects by itself, when combined with sunitinib resulted in delayed resistance in vivo accompanied by a prolonged reduction of microvascular density and tumor perfusion as measured by perfusion imaging relative to sunitinib alone. Sunitinib 98-107 C-X-C motif chemokine ligand 9 Homo sapiens 26-31 20699227-7 2010 Intratumoral injection of CXCL9, although producing minimal effects by itself, when combined with sunitinib resulted in delayed resistance in vivo accompanied by a prolonged reduction of microvascular density and tumor perfusion as measured by perfusion imaging relative to sunitinib alone. Sunitinib 274-283 C-X-C motif chemokine ligand 9 Homo sapiens 26-31 20039326-6 2010 The addition of sunitinib to VEGF-stimulated NCI-N87 cells was associated with a reduction in MAPK phosphorylation (pMAPK) but not Akt phosphorylation (pAkt), whereas the addition of vandetanib was associated with reductions in both VEGF- and EGF-mediated VEGFR2 phosphorylation, pMAPK and pAkt. Sunitinib 16-25 vascular endothelial growth factor A Homo sapiens 29-33 20652398-2 2010 The method involves a two-arm open-label (2:1 randomization) multicenter, randomized phase II trial evaluating the efficacy of sunitinib (arm A) versus no therapy (arm B) in patients with HER-2-negative metastatic breast cancer who achieved an objective response to taxane-based chemotherapy. Sunitinib 127-136 erb-b2 receptor tyrosine kinase 2 Homo sapiens 188-193 20625300-4 2010 Sunitinib, sorafenib, bevacizumab with interferon-alpha, pazopanib, temsirolimus, and everolimus have improved clinical outcomes in randomized phase III clinical trials by inhibiting vascular endothelial growth factors, other tyrosine kinases, the mammalian target of rapamycin pathway, and other related pathways. Sunitinib 0-9 mechanistic target of rapamycin kinase Homo sapiens 248-277 20922699-7 2010 Inhibitors of vascular endothelial growth factor (VEGF), such as bevacizumab, sunitinib, and sorafenib, interfere with angiogenesis. Sunitinib 78-87 vascular endothelial growth factor A Homo sapiens 14-48 20922699-7 2010 Inhibitors of vascular endothelial growth factor (VEGF), such as bevacizumab, sunitinib, and sorafenib, interfere with angiogenesis. Sunitinib 78-87 vascular endothelial growth factor A Homo sapiens 50-54 20922699-9 2010 Sunitinib and sorafenib are small molecules inhibiting tyrosine kinase of the intracellular domain of the VEGF receptor. Sunitinib 0-9 vascular endothelial growth factor A Homo sapiens 106-110 20637791-4 2010 But apoptotic markers (PARP cleavage, caspase 3 cleavage and chromatin condensation) were uniformly negative in H9c2 cells after Sunitinib treatment for 48 h, indicating that another cell death pathway may be involved in Sunitinib-induced cardiotoxicity. Sunitinib 129-138 caspase 3 Rattus norvegicus 38-47 20637791-7 2010 Furthermore, knocking down Beclin 1 by RNA-interference to block autophagy in H9c2 cells revealed that the death rate was decreased when treated with Sunitinib in comparison to control cells. Sunitinib 150-159 beclin 1 Rattus norvegicus 27-35 20659021-6 2010 Our results suggest that H-1152 and sunitinib induce dephosphorylation of Ser910 and Ser935 by inhibiting LRRK2 kinase activity, as these compounds failed to induce significant dephosphorylation of a drug-resistant LRRK2(A2016T) mutant. Sunitinib 36-45 leucine rich repeat kinase 2 Homo sapiens 106-111 20659021-6 2010 Our results suggest that H-1152 and sunitinib induce dephosphorylation of Ser910 and Ser935 by inhibiting LRRK2 kinase activity, as these compounds failed to induce significant dephosphorylation of a drug-resistant LRRK2(A2016T) mutant. Sunitinib 36-45 leucine rich repeat kinase 2 Homo sapiens 215-220 20840755-0 2010 Brain natriuretic peptide precursor (NT-pro-BNP) levels predict for clinical benefit to sunitinib treatment in patients with metastatic renal cell carcinoma. Sunitinib 88-97 natriuretic peptide B Homo sapiens 44-47 20230385-5 2010 RESULTS: There was heterogeneity in tumour responsiveness to sunitinib or sorafenib according to CAIX status; the mean shrinkage was -17% vs -25% for sunitinib-treated patients with high vs low tumour CAIX expression, compared to -13% vs +9% for sorafenib-treated patients (P interaction, 0.05). Sunitinib 61-70 carbonic anhydrase 9 Homo sapiens 97-101 20230385-5 2010 RESULTS: There was heterogeneity in tumour responsiveness to sunitinib or sorafenib according to CAIX status; the mean shrinkage was -17% vs -25% for sunitinib-treated patients with high vs low tumour CAIX expression, compared to -13% vs +9% for sorafenib-treated patients (P interaction, 0.05). Sunitinib 61-70 carbonic anhydrase 9 Homo sapiens 201-205 20230385-5 2010 RESULTS: There was heterogeneity in tumour responsiveness to sunitinib or sorafenib according to CAIX status; the mean shrinkage was -17% vs -25% for sunitinib-treated patients with high vs low tumour CAIX expression, compared to -13% vs +9% for sorafenib-treated patients (P interaction, 0.05). Sunitinib 150-159 carbonic anhydrase 9 Homo sapiens 97-101 20230385-5 2010 RESULTS: There was heterogeneity in tumour responsiveness to sunitinib or sorafenib according to CAIX status; the mean shrinkage was -17% vs -25% for sunitinib-treated patients with high vs low tumour CAIX expression, compared to -13% vs +9% for sorafenib-treated patients (P interaction, 0.05). Sunitinib 150-159 carbonic anhydrase 9 Homo sapiens 201-205 20039326-5 2010 Both EGF and VEGF were shown to stimulate tumor cell growth, and both sunitinib and vandetanib were found to be associated with significant dose-dependent inhibition of proliferation and an enhancement of apoptosis, as determined by MTT and propidium iodide/Annexin V labeling assays, respectively. Sunitinib 70-79 annexin A5 Homo sapiens 258-267 20039326-6 2010 The addition of sunitinib to VEGF-stimulated NCI-N87 cells was associated with a reduction in MAPK phosphorylation (pMAPK) but not Akt phosphorylation (pAkt), whereas the addition of vandetanib was associated with reductions in both VEGF- and EGF-mediated VEGFR2 phosphorylation, pMAPK and pAkt. Sunitinib 16-25 vascular endothelial growth factor A Homo sapiens 233-237 20039326-6 2010 The addition of sunitinib to VEGF-stimulated NCI-N87 cells was associated with a reduction in MAPK phosphorylation (pMAPK) but not Akt phosphorylation (pAkt), whereas the addition of vandetanib was associated with reductions in both VEGF- and EGF-mediated VEGFR2 phosphorylation, pMAPK and pAkt. Sunitinib 16-25 kinase insert domain receptor Homo sapiens 256-262 20133077-13 2010 CONCLUSIONS: These preliminary results suggest that sunitinib and helical tomotherapy yield high Response Evaluation Criteria in Solid Tumors (RECIST) and AFP response rates in advanced HCC with an acceptable safety profile. Sunitinib 52-61 alpha fetoprotein Homo sapiens 155-158 20629553-0 2010 Sunitinib inhibits MEK/ERK and SAPK/JNK pathways and increases sodium/iodide symporter expression in papillary thyroid cancer. Sunitinib 0-9 mitogen-activated protein kinase kinase 7 Homo sapiens 19-22 20629553-1 2010 BACKGROUND: Sunitinib malate (Sutent, Pfizer, Inc.; SU11248) is a selective, multitargeted inhibitor of receptor tyrosine kinases and has been shown to inhibit receptors for VEGF, PDGF, KIT, FLT3, and RET. Sunitinib 12-28 vascular endothelial growth factor A Homo sapiens 174-178 20629553-0 2010 Sunitinib inhibits MEK/ERK and SAPK/JNK pathways and increases sodium/iodide symporter expression in papillary thyroid cancer. Sunitinib 0-9 mitogen-activated protein kinase 1 Homo sapiens 23-26 20629553-1 2010 BACKGROUND: Sunitinib malate (Sutent, Pfizer, Inc.; SU11248) is a selective, multitargeted inhibitor of receptor tyrosine kinases and has been shown to inhibit receptors for VEGF, PDGF, KIT, FLT3, and RET. Sunitinib 12-28 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 186-189 20629553-1 2010 BACKGROUND: Sunitinib malate (Sutent, Pfizer, Inc.; SU11248) is a selective, multitargeted inhibitor of receptor tyrosine kinases and has been shown to inhibit receptors for VEGF, PDGF, KIT, FLT3, and RET. Sunitinib 12-28 fms related receptor tyrosine kinase 3 Homo sapiens 191-195 20629553-0 2010 Sunitinib inhibits MEK/ERK and SAPK/JNK pathways and increases sodium/iodide symporter expression in papillary thyroid cancer. Sunitinib 0-9 mitogen-activated protein kinase 9 Homo sapiens 31-35 20629553-1 2010 BACKGROUND: Sunitinib malate (Sutent, Pfizer, Inc.; SU11248) is a selective, multitargeted inhibitor of receptor tyrosine kinases and has been shown to inhibit receptors for VEGF, PDGF, KIT, FLT3, and RET. Sunitinib 12-28 ret proto-oncogene Homo sapiens 201-204 20629553-1 2010 BACKGROUND: Sunitinib malate (Sutent, Pfizer, Inc.; SU11248) is a selective, multitargeted inhibitor of receptor tyrosine kinases and has been shown to inhibit receptors for VEGF, PDGF, KIT, FLT3, and RET. Sunitinib 30-36 vascular endothelial growth factor A Homo sapiens 174-178 20629553-1 2010 BACKGROUND: Sunitinib malate (Sutent, Pfizer, Inc.; SU11248) is a selective, multitargeted inhibitor of receptor tyrosine kinases and has been shown to inhibit receptors for VEGF, PDGF, KIT, FLT3, and RET. Sunitinib 30-36 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 186-189 20629553-1 2010 BACKGROUND: Sunitinib malate (Sutent, Pfizer, Inc.; SU11248) is a selective, multitargeted inhibitor of receptor tyrosine kinases and has been shown to inhibit receptors for VEGF, PDGF, KIT, FLT3, and RET. Sunitinib 30-36 fms related receptor tyrosine kinase 3 Homo sapiens 191-195 20629553-0 2010 Sunitinib inhibits MEK/ERK and SAPK/JNK pathways and increases sodium/iodide symporter expression in papillary thyroid cancer. Sunitinib 0-9 mitogen-activated protein kinase 8 Homo sapiens 36-39 20629553-1 2010 BACKGROUND: Sunitinib malate (Sutent, Pfizer, Inc.; SU11248) is a selective, multitargeted inhibitor of receptor tyrosine kinases and has been shown to inhibit receptors for VEGF, PDGF, KIT, FLT3, and RET. Sunitinib 30-36 ret proto-oncogene Homo sapiens 201-204 20686603-3 2010 Sunitinib is a drug targeting receptor tyrosine kinases (RTK) like PDGFRalpha, c-Kit and VEGFR-2. Sunitinib 0-9 platelet derived growth factor receptor alpha Homo sapiens 67-77 20629553-1 2010 BACKGROUND: Sunitinib malate (Sutent, Pfizer, Inc.; SU11248) is a selective, multitargeted inhibitor of receptor tyrosine kinases and has been shown to inhibit receptors for VEGF, PDGF, KIT, FLT3, and RET. Sunitinib 52-59 vascular endothelial growth factor A Homo sapiens 174-178 20629553-1 2010 BACKGROUND: Sunitinib malate (Sutent, Pfizer, Inc.; SU11248) is a selective, multitargeted inhibitor of receptor tyrosine kinases and has been shown to inhibit receptors for VEGF, PDGF, KIT, FLT3, and RET. Sunitinib 52-59 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 186-189 20629553-1 2010 BACKGROUND: Sunitinib malate (Sutent, Pfizer, Inc.; SU11248) is a selective, multitargeted inhibitor of receptor tyrosine kinases and has been shown to inhibit receptors for VEGF, PDGF, KIT, FLT3, and RET. Sunitinib 52-59 fms related receptor tyrosine kinase 3 Homo sapiens 191-195 20629553-1 2010 BACKGROUND: Sunitinib malate (Sutent, Pfizer, Inc.; SU11248) is a selective, multitargeted inhibitor of receptor tyrosine kinases and has been shown to inhibit receptors for VEGF, PDGF, KIT, FLT3, and RET. Sunitinib 52-59 ret proto-oncogene Homo sapiens 201-204 20629553-2 2010 The objective of this study was to determine the effects of sunitinib on signal transduction pathways and on gene expression of iodide-metabolizing proteins in papillary cancer cells with the RET/PTC1 rearrangement. Sunitinib 60-69 ret proto-oncogene Homo sapiens 192-195 20629553-2 2010 The objective of this study was to determine the effects of sunitinib on signal transduction pathways and on gene expression of iodide-metabolizing proteins in papillary cancer cells with the RET/PTC1 rearrangement. Sunitinib 60-69 patched 1 Homo sapiens 196-200 20629553-4 2010 RESULTS: Sunitinib inhibited proliferation of RET/PTC1 subclones in a time- and dose-related manner. Sunitinib 9-18 ret proto-oncogene Homo sapiens 46-49 20629553-4 2010 RESULTS: Sunitinib inhibited proliferation of RET/PTC1 subclones in a time- and dose-related manner. Sunitinib 9-18 patched 1 Homo sapiens 50-54 20629553-6 2010 Incubation of RET/PTC1 cells with 1 microM sunitinib inhibited their migration potential and transformed their morphology. Sunitinib 43-52 ret proto-oncogene Homo sapiens 14-17 20629553-6 2010 Incubation of RET/PTC1 cells with 1 microM sunitinib inhibited their migration potential and transformed their morphology. Sunitinib 43-52 patched 1 Homo sapiens 18-22 20629553-7 2010 Sunitinib inhibited RET autophosphorylation at Y1062 and the activation of signal transducer and activator of transcription 3 by blocking Y705 phosphorylation. Sunitinib 0-9 ret proto-oncogene Homo sapiens 20-23 20629553-7 2010 Sunitinib inhibited RET autophosphorylation at Y1062 and the activation of signal transducer and activator of transcription 3 by blocking Y705 phosphorylation. Sunitinib 0-9 signal transducer and activator of transcription 3 Homo sapiens 75-125 20629553-9 2010 Western blot analysis of the p38, MEK/ERK, and SAPK/JNK mitogen-activated protein kinase signal transduction pathways showed that sunitinib blocked ERK 1/2 and JNK phosphorylation in the cytoplasm. Sunitinib 130-139 mitogen-activated protein kinase 1 Homo sapiens 29-32 20629553-9 2010 Western blot analysis of the p38, MEK/ERK, and SAPK/JNK mitogen-activated protein kinase signal transduction pathways showed that sunitinib blocked ERK 1/2 and JNK phosphorylation in the cytoplasm. Sunitinib 130-139 mitogen-activated protein kinase kinase 7 Homo sapiens 34-37 20629553-9 2010 Western blot analysis of the p38, MEK/ERK, and SAPK/JNK mitogen-activated protein kinase signal transduction pathways showed that sunitinib blocked ERK 1/2 and JNK phosphorylation in the cytoplasm. Sunitinib 130-139 mitogen-activated protein kinase 1 Homo sapiens 38-41 20629553-9 2010 Western blot analysis of the p38, MEK/ERK, and SAPK/JNK mitogen-activated protein kinase signal transduction pathways showed that sunitinib blocked ERK 1/2 and JNK phosphorylation in the cytoplasm. Sunitinib 130-139 mitogen-activated protein kinase 9 Homo sapiens 47-51 20629553-9 2010 Western blot analysis of the p38, MEK/ERK, and SAPK/JNK mitogen-activated protein kinase signal transduction pathways showed that sunitinib blocked ERK 1/2 and JNK phosphorylation in the cytoplasm. Sunitinib 130-139 mitogen-activated protein kinase 8 Homo sapiens 52-55 20629553-9 2010 Western blot analysis of the p38, MEK/ERK, and SAPK/JNK mitogen-activated protein kinase signal transduction pathways showed that sunitinib blocked ERK 1/2 and JNK phosphorylation in the cytoplasm. Sunitinib 130-139 mitogen-activated protein kinase 3 Homo sapiens 148-155 20629553-9 2010 Western blot analysis of the p38, MEK/ERK, and SAPK/JNK mitogen-activated protein kinase signal transduction pathways showed that sunitinib blocked ERK 1/2 and JNK phosphorylation in the cytoplasm. Sunitinib 130-139 mitogen-activated protein kinase 8 Homo sapiens 160-163 20629553-10 2010 Sunitinib treatment of RET/PTC1 cell lines, in combination, with forskolin induced expression of the sodium (Na)/iodide (I) symporter (NIS) and the transcription factors that bind the NIS upstream enhancer. Sunitinib 0-9 ret proto-oncogene Homo sapiens 23-26 20629553-10 2010 Sunitinib treatment of RET/PTC1 cell lines, in combination, with forskolin induced expression of the sodium (Na)/iodide (I) symporter (NIS) and the transcription factors that bind the NIS upstream enhancer. Sunitinib 0-9 patched 1 Homo sapiens 27-31 20629553-12 2010 CONCLUSION: Sunitinib appears to target the cytosolic MEK/ERK and SAPK/JNK pathways in the RET/PTC1 cell lines, suggesting that blocking these pathways is at least part of the mechanism by which sunitinib inhibits cell proliferation and causes stimulation of NIS gene expression in RET/PTC1 cells. Sunitinib 12-21 mitogen-activated protein kinase kinase 7 Homo sapiens 54-57 20629553-12 2010 CONCLUSION: Sunitinib appears to target the cytosolic MEK/ERK and SAPK/JNK pathways in the RET/PTC1 cell lines, suggesting that blocking these pathways is at least part of the mechanism by which sunitinib inhibits cell proliferation and causes stimulation of NIS gene expression in RET/PTC1 cells. Sunitinib 12-21 mitogen-activated protein kinase 1 Homo sapiens 58-61 20629553-12 2010 CONCLUSION: Sunitinib appears to target the cytosolic MEK/ERK and SAPK/JNK pathways in the RET/PTC1 cell lines, suggesting that blocking these pathways is at least part of the mechanism by which sunitinib inhibits cell proliferation and causes stimulation of NIS gene expression in RET/PTC1 cells. Sunitinib 12-21 mitogen-activated protein kinase 9 Homo sapiens 66-70 20629553-12 2010 CONCLUSION: Sunitinib appears to target the cytosolic MEK/ERK and SAPK/JNK pathways in the RET/PTC1 cell lines, suggesting that blocking these pathways is at least part of the mechanism by which sunitinib inhibits cell proliferation and causes stimulation of NIS gene expression in RET/PTC1 cells. Sunitinib 12-21 mitogen-activated protein kinase 8 Homo sapiens 71-74 20629553-12 2010 CONCLUSION: Sunitinib appears to target the cytosolic MEK/ERK and SAPK/JNK pathways in the RET/PTC1 cell lines, suggesting that blocking these pathways is at least part of the mechanism by which sunitinib inhibits cell proliferation and causes stimulation of NIS gene expression in RET/PTC1 cells. Sunitinib 12-21 ret proto-oncogene Homo sapiens 91-94 20629553-12 2010 CONCLUSION: Sunitinib appears to target the cytosolic MEK/ERK and SAPK/JNK pathways in the RET/PTC1 cell lines, suggesting that blocking these pathways is at least part of the mechanism by which sunitinib inhibits cell proliferation and causes stimulation of NIS gene expression in RET/PTC1 cells. Sunitinib 12-21 patched 1 Homo sapiens 95-99 20629553-12 2010 CONCLUSION: Sunitinib appears to target the cytosolic MEK/ERK and SAPK/JNK pathways in the RET/PTC1 cell lines, suggesting that blocking these pathways is at least part of the mechanism by which sunitinib inhibits cell proliferation and causes stimulation of NIS gene expression in RET/PTC1 cells. Sunitinib 12-21 ret proto-oncogene Homo sapiens 282-285 20629553-12 2010 CONCLUSION: Sunitinib appears to target the cytosolic MEK/ERK and SAPK/JNK pathways in the RET/PTC1 cell lines, suggesting that blocking these pathways is at least part of the mechanism by which sunitinib inhibits cell proliferation and causes stimulation of NIS gene expression in RET/PTC1 cells. Sunitinib 12-21 patched 1 Homo sapiens 286-290 20013807-9 2010 However, imatinib and sunitinib did induce secretion of anti-inflammatory IL-10 in macrophage cultures, indicating that treatment with these inhibitors might contribute to an immune suppressive microenvironment in GIST. Sunitinib 22-31 interleukin 10 Homo sapiens 74-79 20545945-2 2010 By applying high-throughput protein engineering and crystallization, we have determined the X-ray crystal structures of IL-2-inducible T cell kinase in complex with its selective inhibitor BMS-509744 and the broad-spectrum kinase inhibitors sunitinib and RO5191614. Sunitinib 241-250 interleukin 2 Homo sapiens 120-124 20545945-3 2010 Sunitinib uniquely stabilizes IL-2-inducible T cell kinase in the helix C-in conformation by inducing side chain conformational changes in the ATP-binding site. Sunitinib 0-9 IL2 inducible T cell kinase Homo sapiens 30-58 19547919-2 2010 We conducted a single-institution phase II trial of sunitinib, an oral inhibitor of the VEGF receptor, in patients with relapsed or refractory GCT. Sunitinib 52-61 vascular endothelial growth factor A Homo sapiens 88-92 20520641-0 2010 Identification of a secondary FLT3/A848P mutation in a patient with FLT3-ITD-positive blast phase CMML and response to sunitinib and sorafenib. Sunitinib 119-128 fms related receptor tyrosine kinase 3 Homo sapiens 30-34 20520641-0 2010 Identification of a secondary FLT3/A848P mutation in a patient with FLT3-ITD-positive blast phase CMML and response to sunitinib and sorafenib. Sunitinib 119-128 fms related receptor tyrosine kinase 3 Homo sapiens 68-72 20179017-9 2010 Our study is the first to show a direct link between MGMT expression and decreased angiogenicity and tumorigenicity of GBM cells and suggests the combination of sunitinib and standard therapy as an alternative strategy for GBM patients with MGMT-positive tumors. Sunitinib 161-170 O-6-methylguanine-DNA methyltransferase Homo sapiens 53-57 20137855-1 2010 The present data showed that sunitinib potentiated the in vitro and in vivo anticancer capabilities of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), also known as Apo2 ligand. Sunitinib 29-38 TNF superfamily member 10 Homo sapiens 103-158 20137855-1 2010 The present data showed that sunitinib potentiated the in vitro and in vivo anticancer capabilities of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), also known as Apo2 ligand. Sunitinib 29-38 TNF superfamily member 10 Homo sapiens 160-165 20137855-1 2010 The present data showed that sunitinib potentiated the in vitro and in vivo anticancer capabilities of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), also known as Apo2 ligand. Sunitinib 29-38 TNF superfamily member 10 Homo sapiens 182-193 20163917-5 2010 In contrast, non-VEGF-related side effects are observed with agents inhibiting multiple receptor tyrosine kinases (sunitinib, sorafenib, axitinib and pazopanib) and mammalian target of rapamycin inhibitors (temsirolimus and everolimus); these include diarrhoea, skin rash, stomatitis, hand-foot skin reaction, hypothyroidism, and haematological and metabolic abnormalities. Sunitinib 115-124 vascular endothelial growth factor A Homo sapiens 17-21 20179017-0 2010 MGMT modulates glioblastoma angiogenesis and response to the tyrosine kinase inhibitor sunitinib. Sunitinib 87-96 O-6-methylguanine-DNA methyltransferase Homo sapiens 0-4 20179017-3 2010 We showed that the addition of sunitinib to standard therapy (temozolomide [TMZ] and radiation therapy [RT]) significantly improved the response of MGMT-positive but not of MGMT-negative cells. Sunitinib 31-40 O-6-methylguanine-DNA methyltransferase Homo sapiens 148-152 20179017-3 2010 We showed that the addition of sunitinib to standard therapy (temozolomide [TMZ] and radiation therapy [RT]) significantly improved the response of MGMT-positive but not of MGMT-negative cells. Sunitinib 31-40 O-6-methylguanine-DNA methyltransferase Homo sapiens 173-177 20137855-6 2010 Furthermore, treatment of colon cancer SW620-bearing nude mice with sunitinib plus TRAIL resulted in more significant tumor growth inhibition (52.8%), comparing with the moderate inhibition in TRAIL-treated (35.3%) or sunitinib-treated groups (26.7%) (p<0.05). Sunitinib 68-77 tumor necrosis factor (ligand) superfamily, member 10 Mus musculus 193-198 20137855-6 2010 Furthermore, treatment of colon cancer SW620-bearing nude mice with sunitinib plus TRAIL resulted in more significant tumor growth inhibition (52.8%), comparing with the moderate inhibition in TRAIL-treated (35.3%) or sunitinib-treated groups (26.7%) (p<0.05). Sunitinib 218-227 tumor necrosis factor (ligand) superfamily, member 10 Mus musculus 83-88 20686603-3 2010 Sunitinib is a drug targeting receptor tyrosine kinases (RTK) like PDGFRalpha, c-Kit and VEGFR-2. Sunitinib 0-9 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 79-84 20686603-3 2010 Sunitinib is a drug targeting receptor tyrosine kinases (RTK) like PDGFRalpha, c-Kit and VEGFR-2. Sunitinib 0-9 kinase insert domain receptor Homo sapiens 89-96 20599738-0 2010 Sunitinib deregulates tumor adaptation to hypoxia by inhibiting HIF-1alpha synthesis in HT-29 colon cancer cells. Sunitinib 0-9 hypoxia inducible factor 1 subunit alpha Homo sapiens 64-74 20686603-10 2010 Moreover, sunitinib induced apoptosis and prevented PDGF-AA induced signaling via PDGFRalpha as determined by western blotting. Sunitinib 10-19 platelet derived growth factor receptor alpha Homo sapiens 82-92 20599738-2 2010 Preclinical studies demonstrated that the sunitinib effects are attributed to inhibition of VEGFR and PDGFR phosphorylation. Sunitinib 42-51 kinase insert domain receptor Homo sapiens 92-97 20599738-2 2010 Preclinical studies demonstrated that the sunitinib effects are attributed to inhibition of VEGFR and PDGFR phosphorylation. Sunitinib 42-51 platelet derived growth factor receptor beta Homo sapiens 102-107 20599738-5 2010 First we found that sunitinib limits the colony growth of HT-29, which is a colon adenocarcinoma cell line lacking the RTKs, and that HIF-1alpha in the colonies is decreased by sunitinib. Sunitinib 177-186 hypoxia inducible factor 1 subunit alpha Homo sapiens 134-144 20686603-12 2010 We show that VEGF triggered signal transduction via the MAPK pathway, which could be blocked by sunitinib. Sunitinib 96-105 vascular endothelial growth factor A Homo sapiens 13-17 20599738-6 2010 In cultured HT-29 cells, sunitinib suppressed HIF-1alpha under hypoxic conditions. Sunitinib 25-34 hypoxia inducible factor 1 subunit alpha Homo sapiens 46-56 20599738-7 2010 Moreover, sunitinib repressed the activity of HIF-1alpha and subsequently decreased the expressions of HIF-1 downstream genes. Sunitinib 10-19 hypoxia inducible factor 1 subunit alpha Homo sapiens 46-56 20686603-14 2010 Presence of the 4q12 amplicon and subsequent over-expression of PDGFRA might serve as predictive markers for efficacy of sunitinib. Sunitinib 121-130 platelet derived growth factor receptor alpha Homo sapiens 64-70 20599738-7 2010 Moreover, sunitinib repressed the activity of HIF-1alpha and subsequently decreased the expressions of HIF-1 downstream genes. Sunitinib 10-19 hypoxia inducible factor 1 subunit alpha Homo sapiens 46-51 20599738-8 2010 Mechanistically, sunitinib blocked the 5"-UTR-dependent translation of HIF-1alpha. Sunitinib 17-26 hypoxia inducible factor 1 subunit alpha Homo sapiens 71-81 20601807-7 2010 Sunitinib, which targets VEGF-1, 2 and 3 and PDGF seems to be a well tolerated treatment for advanced tumors. Sunitinib 0-9 vascular endothelial growth factor A Homo sapiens 25-29 20599738-9 2010 The HIF-1alpha suppression by sunitinib was also reproduced in a VHL-null renal cell carcinoma cell line, where HIF-1alpha is not degradable. Sunitinib 30-39 hypoxia inducible factor 1 subunit alpha Homo sapiens 4-14 20599738-9 2010 The HIF-1alpha suppression by sunitinib was also reproduced in a VHL-null renal cell carcinoma cell line, where HIF-1alpha is not degradable. Sunitinib 30-39 hypoxia inducible factor 1 subunit alpha Homo sapiens 112-122 20599738-10 2010 In conclusion, the sunitinib inhibition of HIF-1 signaling could restrain tumor progression in hypoxic regions, which may contribute to anticancer effect of sunitinib. Sunitinib 19-28 hypoxia inducible factor 1 subunit alpha Homo sapiens 43-48 20599738-10 2010 In conclusion, the sunitinib inhibition of HIF-1 signaling could restrain tumor progression in hypoxic regions, which may contribute to anticancer effect of sunitinib. Sunitinib 157-166 hypoxia inducible factor 1 subunit alpha Homo sapiens 43-48 20558072-4 2010 Remarkably, dimethylation of both the 4-N and N7 afford whole cell EGFR inhibitors that are more cytotoxic than clinically used erlotinib and mono-methylation at the 4-N or N7 affords more cytotoxic whole cell PDGFR-beta inhibitors than clinically used sunitinib. Sunitinib 253-262 epidermal growth factor receptor Homo sapiens 67-71 20204363-3 2010 Sunitinib malate is a VEGFR inhibitor approved for the treatment of advanced renal cell carcinoma. Sunitinib 0-16 kinase insert domain receptor Homo sapiens 22-27 20345483-0 2010 Pharmacological interaction with sunitinib is abolished by a germ-line mutation (1291T>C) of BCRP/ABCG2 gene. Sunitinib 33-42 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 96-100 20495370-0 2010 SU11248 (sunitinib) directly inhibits the activity of mammalian 5"-AMP-activated protein kinase (AMPK). Sunitinib 0-7 protein kinase AMP-activated catalytic subunit alpha 2 Homo sapiens 97-101 20495370-0 2010 SU11248 (sunitinib) directly inhibits the activity of mammalian 5"-AMP-activated protein kinase (AMPK). Sunitinib 9-18 protein kinase AMP-activated catalytic subunit alpha 2 Homo sapiens 97-101 20495370-6 2010 Here we report that the chemotherapeutic drug SU11248 (sunitinib) is at least as potent an inhibitor of AMPK as compound C, which is a commonly used experimental direct inhibitor of the enzyme. Sunitinib 46-53 protein kinase AMP-activated catalytic subunit alpha 2 Homo sapiens 104-108 20495370-6 2010 Here we report that the chemotherapeutic drug SU11248 (sunitinib) is at least as potent an inhibitor of AMPK as compound C, which is a commonly used experimental direct inhibitor of the enzyme. Sunitinib 55-64 protein kinase AMP-activated catalytic subunit alpha 2 Homo sapiens 104-108 20495370-8 2010 The ability of SU11248 to inhibit AMPK has potential clinical significance--there may be populations of SU11248-treated patients in which AMPK activity is inhibited in normal as well as in tumor tissue. Sunitinib 15-22 protein kinase AMP-activated catalytic subunit alpha 2 Homo sapiens 34-38 20495370-8 2010 The ability of SU11248 to inhibit AMPK has potential clinical significance--there may be populations of SU11248-treated patients in which AMPK activity is inhibited in normal as well as in tumor tissue. Sunitinib 15-22 protein kinase AMP-activated catalytic subunit alpha 2 Homo sapiens 138-142 20495370-8 2010 The ability of SU11248 to inhibit AMPK has potential clinical significance--there may be populations of SU11248-treated patients in which AMPK activity is inhibited in normal as well as in tumor tissue. Sunitinib 104-111 protein kinase AMP-activated catalytic subunit alpha 2 Homo sapiens 34-38 20495370-8 2010 The ability of SU11248 to inhibit AMPK has potential clinical significance--there may be populations of SU11248-treated patients in which AMPK activity is inhibited in normal as well as in tumor tissue. Sunitinib 104-111 protein kinase AMP-activated catalytic subunit alpha 2 Homo sapiens 138-142 20571071-4 2010 Small molecule tyrosine kinase inhibitors (TKI) targeting the VEGF receptor (i.e., sunitinib, sorafenib, and vandetanib) show activity in phase II clinical studies. Sunitinib 83-92 vascular endothelial growth factor A Homo sapiens 62-66 20509775-2 2010 The efforts in this area have been greatly enhanced by the approval of tyrosine kinase inhibitors with PDGFR inhibitory activity such as imatinib, sunitinib and sorafenib. Sunitinib 147-156 platelet derived growth factor receptor beta Homo sapiens 103-108 20533592-4 2010 Imatinib and sunitinib, both tyrosine kinase inhibitors directed to KIT, were approved for first- and second-line treatment of metastatic and unresectable GISTs. Sunitinib 13-22 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 68-71 20345483-0 2010 Pharmacological interaction with sunitinib is abolished by a germ-line mutation (1291T>C) of BCRP/ABCG2 gene. Sunitinib 33-42 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 101-106 20345483-4 2010 Sunitinib remarkably reversed BCRP-mediated and partially reversed P-gp-mediated drug resistance in the respective transfectants. Sunitinib 0-9 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 30-34 20345483-4 2010 Sunitinib remarkably reversed BCRP-mediated and partially reversed P-gp-mediated drug resistance in the respective transfectants. Sunitinib 0-9 ATP binding cassette subfamily B member 1 Homo sapiens 67-71 20345483-5 2010 The in vitro vesicle transport assay indicated that sunitinib competitively inhibited BCRP-mediated estrone 3-sulfate transport and P-gp-mediated vincristine transport. Sunitinib 52-61 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 86-90 20345483-5 2010 The in vitro vesicle transport assay indicated that sunitinib competitively inhibited BCRP-mediated estrone 3-sulfate transport and P-gp-mediated vincristine transport. Sunitinib 52-61 ATP binding cassette subfamily B member 1 Homo sapiens 132-136 20345483-9 2010 Thus, residue Phe-431 of BCRP is important for the pharmacological interaction with sunitinib and FTC. Sunitinib 84-93 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 25-29 19444655-1 2010 Sunitinib is a recently approved tyrosine-kinase inhibitor that targets the vascular endothelial growth factor receptors (VEGFR). Sunitinib 0-9 kinase insert domain receptor Homo sapiens 76-120 20733555-4 2010 Sorafenib and sunitinib were the first FLT3 inhibitors to be studied in the clinic and have the most clinically relevant data. Sunitinib 14-23 fms related receptor tyrosine kinase 3 Homo sapiens 39-43 20484434-0 2010 Molecular imaging of changes in the prevalence of vascular endothelial growth factor receptor in sunitinib-treated murine mammary tumors. Sunitinib 97-106 vascular endothelial growth factor A Mus musculus 50-84 19444655-1 2010 Sunitinib is a recently approved tyrosine-kinase inhibitor that targets the vascular endothelial growth factor receptors (VEGFR). Sunitinib 0-9 kinase insert domain receptor Homo sapiens 122-127 20067776-7 2010 Many of these drugs, including emetine, fluorosalan, sunitinib malate, bithionol, narasin, tribromsalan, and lestaurtinib, inhibited NF-kappaB signaling via inhibition of IkappaBalpha phosphorylation. Sunitinib 53-69 nuclear factor kappa B subunit 1 Homo sapiens 133-142 20231424-2 2010 We studied whether sunitinib, a multityrosine kinase inhibitor that inhibits KIT, enhances engraftment after bone marrow transplantation (BMT) in mice. Sunitinib 19-28 KIT proto-oncogene receptor tyrosine kinase Mus musculus 77-80 20231424-7 2010 Sunitinib did not increase engraftment in mice with deficient KIT signaling, and the pattern of more potent effects on T cell compared with HSC engraftment observed in sunitinib-treated hosts was also observed after BMT into KIT(W/Wv) mice. Sunitinib 168-177 KIT proto-oncogene receptor tyrosine kinase Mus musculus 225-228 20502715-0 2010 Soluble isoforms of vascular endothelial growth factor are predictors of response to sunitinib in metastatic renal cell carcinomas. Sunitinib 85-94 vascular endothelial growth factor A Homo sapiens 20-54 20502715-8 2010 Among transcripts analyzed, only the levels of vascular endothelial growth factor (VEGF) soluble isoforms (VEGF(121) and VEGF(165)) were associated with the response to sunitinib (P = 0.04 for both). Sunitinib 169-178 vascular endothelial growth factor A Homo sapiens 47-81 20502715-8 2010 Among transcripts analyzed, only the levels of vascular endothelial growth factor (VEGF) soluble isoforms (VEGF(121) and VEGF(165)) were associated with the response to sunitinib (P = 0.04 for both). Sunitinib 169-178 vascular endothelial growth factor A Homo sapiens 83-87 20502715-8 2010 Among transcripts analyzed, only the levels of vascular endothelial growth factor (VEGF) soluble isoforms (VEGF(121) and VEGF(165)) were associated with the response to sunitinib (P = 0.04 for both). Sunitinib 169-178 vascular endothelial growth factor A Homo sapiens 107-111 20502715-8 2010 Among transcripts analyzed, only the levels of vascular endothelial growth factor (VEGF) soluble isoforms (VEGF(121) and VEGF(165)) were associated with the response to sunitinib (P = 0.04 for both). Sunitinib 169-178 vascular endothelial growth factor A Homo sapiens 107-111 20208561-9 2010 In a neuroblastoma xenograft model, tumor growth inhibition by sunitinib was associated with inhibition of angiogenesis and reduced HIF-1alpha levels. Sunitinib 63-72 hypoxia inducible factor 1 subunit alpha Homo sapiens 132-142 20067776-7 2010 Many of these drugs, including emetine, fluorosalan, sunitinib malate, bithionol, narasin, tribromsalan, and lestaurtinib, inhibited NF-kappaB signaling via inhibition of IkappaBalpha phosphorylation. Sunitinib 53-69 NFKB inhibitor alpha Homo sapiens 171-183 20147887-7 2010 Our study shows that serum levels of VEGF and NGAL are significant predictors of progression-free survival in patients with renal cell carcinoma treated with sunitinib. Sunitinib 158-167 lipocalin 2 Homo sapiens 46-50 20406969-6 2010 Proteomic analyses comparing tumor to peripheral compartments showed that granulocyte macrophage colony-stimulating factor (GM-CSF) predicted sunitinib resistance and recombinant GM-CSF conferred sunitinib resistance to MDSC in vivo and in vitro. Sunitinib 142-151 colony stimulating factor 2 Homo sapiens 74-122 20406969-6 2010 Proteomic analyses comparing tumor to peripheral compartments showed that granulocyte macrophage colony-stimulating factor (GM-CSF) predicted sunitinib resistance and recombinant GM-CSF conferred sunitinib resistance to MDSC in vivo and in vitro. Sunitinib 142-151 colony stimulating factor 2 Homo sapiens 124-130 20406969-6 2010 Proteomic analyses comparing tumor to peripheral compartments showed that granulocyte macrophage colony-stimulating factor (GM-CSF) predicted sunitinib resistance and recombinant GM-CSF conferred sunitinib resistance to MDSC in vivo and in vitro. Sunitinib 196-205 colony stimulating factor 2 Homo sapiens 179-185 20406969-8 2010 STAT5ab(null/null) MDSC were rendered sensitive to sunitinib in the presence of GM-CSF in vitro. Sunitinib 51-60 signal transducer and activator of transcription 5A Homo sapiens 0-5 20406969-8 2010 STAT5ab(null/null) MDSC were rendered sensitive to sunitinib in the presence of GM-CSF in vitro. Sunitinib 51-60 colony stimulating factor 2 Homo sapiens 80-86 20406969-9 2010 We conclude that compartment-dependent GM-CSF exposure in resistant tumors may account for the regionalized effect of sunitinib upon host MDSC modulation and hypothesize that ancillary strategies to decrease such regionalized escape will enhance the potency of sunitinib as an immunomodulator and a cancer therapy. Sunitinib 118-127 colony stimulating factor 2 Homo sapiens 39-45 20406969-9 2010 We conclude that compartment-dependent GM-CSF exposure in resistant tumors may account for the regionalized effect of sunitinib upon host MDSC modulation and hypothesize that ancillary strategies to decrease such regionalized escape will enhance the potency of sunitinib as an immunomodulator and a cancer therapy. Sunitinib 261-270 colony stimulating factor 2 Homo sapiens 39-45 20335444-7 2010 VEGF receptor (VEGFR) tyrosine kinase inhibition (through exposure to sunitinib and 676475) blocked vhl(-/-)-induced angiogenesis in all affected tissues, demonstrating that Vegfaa, Vegfab and Vegfb are key effectors of the vhl(-/-) angiogenic phenotype through Flt1, Kdr and Kdr-like signaling. Sunitinib 70-79 vascular endothelial growth factor Aa Danio rerio 0-4 20335444-7 2010 VEGF receptor (VEGFR) tyrosine kinase inhibition (through exposure to sunitinib and 676475) blocked vhl(-/-)-induced angiogenesis in all affected tissues, demonstrating that Vegfaa, Vegfab and Vegfb are key effectors of the vhl(-/-) angiogenic phenotype through Flt1, Kdr and Kdr-like signaling. Sunitinib 70-79 von Hippel-Lindau tumor suppressor Danio rerio 100-103 20351323-1 2010 PURPOSE: Sunitinib and sorafenib are oral vascular endothelial growth factor receptor (VEGFR) tyrosine kinase inhibitors (TKIs) used in a vast range of cancers. Sunitinib 9-18 kinase insert domain receptor Homo sapiens 42-85 20351323-1 2010 PURPOSE: Sunitinib and sorafenib are oral vascular endothelial growth factor receptor (VEGFR) tyrosine kinase inhibitors (TKIs) used in a vast range of cancers. Sunitinib 9-18 kinase insert domain receptor Homo sapiens 87-92 20351323-11 2010 CONCLUSION: Treatment with VEGFR TKIs sunitinib and sorafenib is associated with a significant increase in the risk of ATEs. Sunitinib 38-47 kinase insert domain receptor Homo sapiens 27-32 20147887-0 2010 Predictive value of baseline serum vascular endothelial growth factor and neutrophil gelatinase-associated lipocalin in advanced kidney cancer patients receiving sunitinib. Sunitinib 162-171 vascular endothelial growth factor A Homo sapiens 35-69 20147887-0 2010 Predictive value of baseline serum vascular endothelial growth factor and neutrophil gelatinase-associated lipocalin in advanced kidney cancer patients receiving sunitinib. Sunitinib 162-171 lipocalin 2 Homo sapiens 74-116 20147887-7 2010 Our study shows that serum levels of VEGF and NGAL are significant predictors of progression-free survival in patients with renal cell carcinoma treated with sunitinib. Sunitinib 158-167 vascular endothelial growth factor A Homo sapiens 37-41 20339913-0 2010 Phase III randomized trial of sunitinib versus capecitabine in patients with previously treated HER2-negative advanced breast cancer. Sunitinib 30-39 erb-b2 receptor tyrosine kinase 2 Homo sapiens 96-100 20064605-5 2010 The molecularly targeted cancer therapeutics, sunitinib and imatinib, exhibited profound effects on Ca(i)(2+), combining effects of cytotoxic chemotherapeutics, TdP inducers and potent hERG channel blockers. Sunitinib 46-55 ETS transcription factor ERG Homo sapiens 185-189 20234089-6 2010 However, preconditioning by short-term Sm treatment proved to be successful as an adjunct therapy, increasing the frequency of IFN-gamma+ and IFN-gamma+TNF+CD4+ T cells, enhancing NO production by splenic macrophages, and providing dose-sparing effects when combined with a first-line immune-dependent anti-leishmanial drug. Sunitinib 39-41 interferon gamma Mus musculus 127-136 20234089-6 2010 However, preconditioning by short-term Sm treatment proved to be successful as an adjunct therapy, increasing the frequency of IFN-gamma+ and IFN-gamma+TNF+CD4+ T cells, enhancing NO production by splenic macrophages, and providing dose-sparing effects when combined with a first-line immune-dependent anti-leishmanial drug. Sunitinib 39-41 interferon gamma Mus musculus 142-151 20234089-6 2010 However, preconditioning by short-term Sm treatment proved to be successful as an adjunct therapy, increasing the frequency of IFN-gamma+ and IFN-gamma+TNF+CD4+ T cells, enhancing NO production by splenic macrophages, and providing dose-sparing effects when combined with a first-line immune-dependent anti-leishmanial drug. Sunitinib 39-41 tumor necrosis factor Mus musculus 152-155 20368728-5 2010 Drugs that modulate the downstream targets of the pVHL/HIF pathway, including sunitinib, sorafenib, temsirolimus and bevacizumab, have proven benefit in treating ccRCC. Sunitinib 78-87 von Hippel-Lindau tumor suppressor Homo sapiens 50-54 20015695-5 2010 In addition, sunitinib led to accumulation in G(0)/G(1) phase of the cell cycle, inhibition of cytokine production, downregulation of activation markers expression and blockade of Zap-70 signaling in the T cells. Sunitinib 13-22 zeta chain of T cell receptor associated protein kinase 70 Homo sapiens 180-186 20446917-4 2010 In these conditions, abrogating specifically Pgp-mediated efflux in vitro revealed the remarkable and statistically significant cellular accumulation of imatinib (difference in cellular levels between Pgp-expressing and silenced cells, at high and low incubation concentration, respectively: 6.1 and 6.6), dasatinib (4.9 and 5.6), sunitinib (3.7 and 7.3) and sorafenib (1.2 and 1.4), confirming that these TKIs are all substrates of Pgp. Sunitinib 331-340 ATP binding cassette subfamily B member 1 Homo sapiens 45-48 20147452-0 2010 Function of activation loop tyrosine phosphorylation in the mechanism of c-Kit auto-activation and its implication in sunitinib resistance. Sunitinib 118-127 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 73-78 20658715-2 2010 Vascular endothelial growth factor (VEGF) pathway is the principle target for drugs like sunitinib, sorafenib and bevacizumab. Sunitinib 89-98 vascular endothelial growth factor A Homo sapiens 0-34 20658715-2 2010 Vascular endothelial growth factor (VEGF) pathway is the principle target for drugs like sunitinib, sorafenib and bevacizumab. Sunitinib 89-98 vascular endothelial growth factor A Homo sapiens 36-40 20095048-2 2010 Biophysical, biochemical, and structural studies have provided insight into the molecular basis of resistance to the KIT inhibitors, imatinib and sunitinib. Sunitinib 146-155 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 117-120 20095048-3 2010 Here, solution-phase hydrogen/deuterium exchange (HDX) and direct binding mass spectrometry experiments provide a link between static structure models and the dynamic equilibrium of the multiple states of KIT, supporting that sunitinib targets the autoinhibited conformation of WT-KIT. Sunitinib 226-235 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 205-208 20095048-3 2010 Here, solution-phase hydrogen/deuterium exchange (HDX) and direct binding mass spectrometry experiments provide a link between static structure models and the dynamic equilibrium of the multiple states of KIT, supporting that sunitinib targets the autoinhibited conformation of WT-KIT. Sunitinib 226-235 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 281-284 20095048-6 2010 This result correlates with the V560D mutant exhibiting a sensitivity to sunitinib that is less than for WT KIT but greater than for KIT D816H. Sunitinib 73-82 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 108-111 19626587-0 2010 Rlip76 transports sunitinib and sorafenib and mediates drug resistance in kidney cancer. Sunitinib 18-27 ralA binding protein 1 Homo sapiens 0-6 19626587-6 2010 Results of these studies show that sorafenib as well as sunitinib are substrates for transport by RLIP76 thus are competitive inhibitors of GS-E transport. Sunitinib 56-65 ralA binding protein 1 Homo sapiens 98-104 20019017-4 2010 The renal biopsy revealed focal segmental glomerulosclerosis (FSGS) in addition to thrombotic microangiopathy (TMA), and the complete syndrome disappeared 6 months after sunitinib withdrawal. Sunitinib 170-179 actinin alpha 4 Homo sapiens 62-66 20019017-5 2010 To our knowledge, this is the first case of FSGS associated to TMA secondary to sunitinib treatment. Sunitinib 80-89 actinin alpha 4 Homo sapiens 44-48 19949796-1 2010 The discovery of activating oncogenic C-KIT and PDGFRA mutations in gastrointestinal stromal tumors (GIST) represented the key for the development of innovative targeted molecular therapy using the tyrosine kinase inhibitors (TKI) Imatinib (Glivec(R)), Sunitinib and other substances. Sunitinib 253-262 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 38-43 19949796-1 2010 The discovery of activating oncogenic C-KIT and PDGFRA mutations in gastrointestinal stromal tumors (GIST) represented the key for the development of innovative targeted molecular therapy using the tyrosine kinase inhibitors (TKI) Imatinib (Glivec(R)), Sunitinib and other substances. Sunitinib 253-262 platelet derived growth factor receptor alpha Homo sapiens 48-54 20103651-0 2010 Interleukin-8 mediates resistance to antiangiogenic agent sunitinib in renal cell carcinoma. Sunitinib 58-67 C-X-C motif chemokine ligand 8 Homo sapiens 0-13 20187785-1 2010 BACKGROUND: Sunitinib is a small molecule that inhibits receptor tyrosine kinases, including the vascular endothelial growth factor receptors, and exhibits antiangiogenic and antitumor activity. Sunitinib 12-21 vascular endothelial growth factor A Homo sapiens 97-131 19952730-2 2010 This is the first report of sorafenib and sunitinib, both small molecule tyrosine kinase inhibitors of vascular endothelial growth factor and platelet-derived growth factor receptors, triggering radiation recall dermatitis. Sunitinib 42-51 vascular endothelial growth factor A Homo sapiens 103-137 20103629-4 2010 Microarray expression profiling and Western blot analysis revealed that among known sunitinib targets, only platelet-derived growth factor receptor-beta and vascular endothelial growth factor receptor-2 (VEGFR-2) were overexpressed in ccRCCs relative to normal tissues. Sunitinib 84-93 platelet derived growth factor receptor beta Homo sapiens 108-152 20103629-4 2010 Microarray expression profiling and Western blot analysis revealed that among known sunitinib targets, only platelet-derived growth factor receptor-beta and vascular endothelial growth factor receptor-2 (VEGFR-2) were overexpressed in ccRCCs relative to normal tissues. Sunitinib 84-93 kinase insert domain receptor Homo sapiens 157-202 20103651-6 2010 Notably, escape coincided with increased secretion of interleukin-8 (IL-8) from tumors into the plasma, and coadministration of an IL-8 neutralizing antibody resensitized tumors to sunitinib treatment. Sunitinib 181-190 C-X-C motif chemokine ligand 8 Homo sapiens 131-135 20103629-4 2010 Microarray expression profiling and Western blot analysis revealed that among known sunitinib targets, only platelet-derived growth factor receptor-beta and vascular endothelial growth factor receptor-2 (VEGFR-2) were overexpressed in ccRCCs relative to normal tissues. Sunitinib 84-93 kinase insert domain receptor Homo sapiens 204-211 20103629-6 2010 In contrast, sunitinib inhibited endothelial cell proliferation and motility at the same concentrations by suppressing VEGFR-2 signaling. Sunitinib 13-22 kinase insert domain receptor Homo sapiens 119-126 20103651-7 2010 In patients who were refractory to sunitinib treatment, IL-8 expression was elevated in ccRCC tumors, supporting the concept that IL-8 levels might predict clinical response to sunitinib. Sunitinib 177-186 C-X-C motif chemokine ligand 8 Homo sapiens 56-60 20103651-7 2010 In patients who were refractory to sunitinib treatment, IL-8 expression was elevated in ccRCC tumors, supporting the concept that IL-8 levels might predict clinical response to sunitinib. Sunitinib 177-186 C-X-C motif chemokine ligand 8 Homo sapiens 130-134 20103651-8 2010 Our results reveal IL-8 as an important contributor to sunitinib resistance in ccRCC and a candidate therapeutic target to reverse acquired or intrinsic resistance to sunitinib in this malignancy. Sunitinib 55-64 C-X-C motif chemokine ligand 8 Homo sapiens 19-23 20103651-8 2010 Our results reveal IL-8 as an important contributor to sunitinib resistance in ccRCC and a candidate therapeutic target to reverse acquired or intrinsic resistance to sunitinib in this malignancy. Sunitinib 167-176 C-X-C motif chemokine ligand 8 Homo sapiens 19-23 19940466-5 2010 Sunitinib, a small molecule blocking intracellular VEGF, KIT, Flt3 and PDGF receptors, which regulate angiogenesis and cell growth, is approved for the treatment of advanced renal cell cancer (RCC) and malignant gastrointestinal stromal tumor. Sunitinib 0-9 vascular endothelial growth factor A Homo sapiens 51-55 20721275-2 2010 These include the immunotherapeutics, alfa-interferon, and interleukin-2, and agents that target the vascular endothelial growth factor receptor (VEGFR) via its tyrosine kinase, such as sorafenib, sunitinib, and pazopanib, or the mammalian target of rapamycin (mTOR), such as temsirolimus and everolimus. Sunitinib 197-206 kinase insert domain receptor Homo sapiens 101-144 21067536-6 2010 While, the role of sunitinib and inhibitors of mammalian target of rapamycin (mTOR) in breast cancer has to be defined. Sunitinib 19-28 mechanistic target of rapamycin kinase Homo sapiens 78-82 19996579-3 2010 Recently 12 cases of metastatic melanoma and KIT-activating mutations have been published to be successfully treated with c-KIT blockers such as imatinib, sunitinib, dasatinib or sorafenib. Sunitinib 155-164 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 45-48 19996579-3 2010 Recently 12 cases of metastatic melanoma and KIT-activating mutations have been published to be successfully treated with c-KIT blockers such as imatinib, sunitinib, dasatinib or sorafenib. Sunitinib 155-164 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 122-127 20696054-6 2010 Genes whose protein products are targeted by the RET inhibitors sunitinib and sorafenib correlated with being amplified and or highly expressed. Sunitinib 64-73 ret proto-oncogene Homo sapiens 49-52 20103668-4 2010 It is likely that the multikinase inhibitors, sorafenib, sunitinib, axitinib, and motesanib, whose targets include VEGFR-2, exert their effects primarily through inhibition of endothelial cells. Sunitinib 57-66 kinase insert domain receptor Homo sapiens 115-122 19740535-2 2010 Sunitinib malate is an oral, multi-targeted tyrosine kinase inhibitor that inhibits receptors for VEGF, c-Kit and platelet-derived growth factor. Sunitinib 0-16 vascular endothelial growth factor A Homo sapiens 98-102 19740535-2 2010 Sunitinib malate is an oral, multi-targeted tyrosine kinase inhibitor that inhibits receptors for VEGF, c-Kit and platelet-derived growth factor. Sunitinib 0-16 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 104-109 20031962-8 2010 However, clinical development of small molecule multi-kinase inhibitors including those targeting vascular endothelial growth factor receptors, such as vandetanib, sunitinib and sorafenib, has not been very successful. Sunitinib 164-173 vascular endothelial growth factor A Homo sapiens 98-132 20165865-6 2010 Although, KIT (CD117) immunohistochemistry is a reliable diagnostic tool in the diagnosis of GIST, KIT-negative GISTs, GISTs showing unusual morphology as well as GISTs which progress during or after treatment with imatinib/sunitinib can be a challenge for pathologists and clinicians. Sunitinib 224-233 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 10-13 19900123-2 2010 The receptor tyrosine kinase (RTK) inhibitor imatinib mesylate has been approved as the first-line choice of therapy for this group of patients, while the RTK inhibitor, sunitinib malate, has been approved for the treatment of GIST after disease progression or intolerance to imatinib. Sunitinib 170-186 ret proto-oncogene Homo sapiens 155-158 20721275-2 2010 These include the immunotherapeutics, alfa-interferon, and interleukin-2, and agents that target the vascular endothelial growth factor receptor (VEGFR) via its tyrosine kinase, such as sorafenib, sunitinib, and pazopanib, or the mammalian target of rapamycin (mTOR), such as temsirolimus and everolimus. Sunitinib 197-206 kinase insert domain receptor Homo sapiens 146-151 20949248-2 2010 Sunitinib is able to inhibit RTKs such as receptors for platelet-derived growth factor (PDGF-R alpha and beta) and for vascular endothelial growth factor (VEGFRs). Sunitinib 0-9 platelet derived growth factor receptor alpha Homo sapiens 88-100 20949248-2 2010 Sunitinib is able to inhibit RTKs such as receptors for platelet-derived growth factor (PDGF-R alpha and beta) and for vascular endothelial growth factor (VEGFRs). Sunitinib 0-9 vascular endothelial growth factor A Homo sapiens 119-153 21158731-4 2010 The initial pessimism about the usefulness of the antiangiogenic therapeutic approach for cancer, derived from the poor results obtained in clinical trials, turned into euphoria after the approvals of the anti-VEGF monoclonal antibody bevacizumab and the multitargeted tyrosine kinase inhibitors sunitinib, sorafenib and pazopanib. Sunitinib 296-305 vascular endothelial growth factor A Homo sapiens 210-214 19940466-5 2010 Sunitinib, a small molecule blocking intracellular VEGF, KIT, Flt3 and PDGF receptors, which regulate angiogenesis and cell growth, is approved for the treatment of advanced renal cell cancer (RCC) and malignant gastrointestinal stromal tumor. Sunitinib 0-9 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 57-60 20102396-9 2010 In vitro, addition of sunitinib prevented the release of endocan in human umbilical vascular endothelial cells when induced by vascular endothelial growth factor. Sunitinib 22-31 endothelial cell specific molecule 1 Homo sapiens 57-64 20102396-9 2010 In vitro, addition of sunitinib prevented the release of endocan in human umbilical vascular endothelial cells when induced by vascular endothelial growth factor. Sunitinib 22-31 vascular endothelial growth factor A Homo sapiens 127-161 19940466-5 2010 Sunitinib, a small molecule blocking intracellular VEGF, KIT, Flt3 and PDGF receptors, which regulate angiogenesis and cell growth, is approved for the treatment of advanced renal cell cancer (RCC) and malignant gastrointestinal stromal tumor. Sunitinib 0-9 fms related receptor tyrosine kinase 3 Homo sapiens 62-66 19833232-0 2010 Therapeutic activity of sunitinib for Her2/neu induced mammary cancer in FVB mice. Sunitinib 24-33 erb-b2 receptor tyrosine kinase 2 Mus musculus 38-42 19833232-0 2010 Therapeutic activity of sunitinib for Her2/neu induced mammary cancer in FVB mice. Sunitinib 24-33 erb-b2 receptor tyrosine kinase 2 Mus musculus 43-46 19833232-5 2010 In association with its therapeutic activity, sunitinib reduced the absolute number of splenic T-reg cells (CD4(+)CD25(+)CD62L(+)) and MDSCs (CD11b(+)Gr1(+)) that were increased during tumor progression with less activity in mice with gross tumors. Sunitinib 46-55 integrin alpha M Mus musculus 142-147 19833232-8 2010 Additionally immune-regulatory cytokines and enzymes were down regulated by sunitinib treatment, including TGFbeta and NOS2 in the spleen cells of sunitinib treated mice as compared to untreated tumor bearing (TB) mice. Sunitinib 76-85 transforming growth factor, beta 1 Mus musculus 107-114 19833232-8 2010 Additionally immune-regulatory cytokines and enzymes were down regulated by sunitinib treatment, including TGFbeta and NOS2 in the spleen cells of sunitinib treated mice as compared to untreated tumor bearing (TB) mice. Sunitinib 76-85 nitric oxide synthase 2, inducible Mus musculus 119-123 19833232-8 2010 Additionally immune-regulatory cytokines and enzymes were down regulated by sunitinib treatment, including TGFbeta and NOS2 in the spleen cells of sunitinib treated mice as compared to untreated tumor bearing (TB) mice. Sunitinib 147-156 transforming growth factor, beta 1 Mus musculus 107-114 19833232-8 2010 Additionally immune-regulatory cytokines and enzymes were down regulated by sunitinib treatment, including TGFbeta and NOS2 in the spleen cells of sunitinib treated mice as compared to untreated tumor bearing (TB) mice. Sunitinib 147-156 nitric oxide synthase 2, inducible Mus musculus 119-123 20215359-3 2010 These include the vascular endothelial growth factor receptor (VEGFR) tyrosine kinase inhibitors sunitinib and sorafenib, which inhibit angiogenic signaling in endothelial cells and vascular pericytes predominantly through VEGFR and platelet-derived growth factor receptor beta. Sunitinib 97-106 kinase insert domain receptor Homo sapiens 18-61 20053726-10 2010 Expression of a constitutively activated STAT3 mutant or myristoylated AKT partially blocked the effects of sunitinib in these tumor cells. Sunitinib 108-117 signal transducer and activator of transcription 3 Homo sapiens 41-46 20053726-10 2010 Expression of a constitutively activated STAT3 mutant or myristoylated AKT partially blocked the effects of sunitinib in these tumor cells. Sunitinib 108-117 AKT serine/threonine kinase 1 Homo sapiens 71-74 20215359-3 2010 These include the vascular endothelial growth factor receptor (VEGFR) tyrosine kinase inhibitors sunitinib and sorafenib, which inhibit angiogenic signaling in endothelial cells and vascular pericytes predominantly through VEGFR and platelet-derived growth factor receptor beta. Sunitinib 97-106 kinase insert domain receptor Homo sapiens 223-228 20215359-3 2010 These include the vascular endothelial growth factor receptor (VEGFR) tyrosine kinase inhibitors sunitinib and sorafenib, which inhibit angiogenic signaling in endothelial cells and vascular pericytes predominantly through VEGFR and platelet-derived growth factor receptor beta. Sunitinib 97-106 platelet derived growth factor receptor beta Homo sapiens 233-277 20053726-0 2010 Sunitinib induces apoptosis and growth arrest of medulloblastoma tumor cells by inhibiting STAT3 and AKT signaling pathways. Sunitinib 0-9 signal transducer and activator of transcription 3 Homo sapiens 91-96 20053726-0 2010 Sunitinib induces apoptosis and growth arrest of medulloblastoma tumor cells by inhibiting STAT3 and AKT signaling pathways. Sunitinib 0-9 AKT serine/threonine kinase 1 Homo sapiens 101-104 20053726-4 2010 Sunitinib treatment resulted in the activation of caspase-3 and cleavage of poly(ADP-ribose) polymerase and upregulation of proapoptotic genes, Bak and Bim, and inhibited the expression of survivin, an antiapoptotic protein. Sunitinib 0-9 caspase 3 Homo sapiens 50-59 20053726-4 2010 Sunitinib treatment resulted in the activation of caspase-3 and cleavage of poly(ADP-ribose) polymerase and upregulation of proapoptotic genes, Bak and Bim, and inhibited the expression of survivin, an antiapoptotic protein. Sunitinib 0-9 poly(ADP-ribose) polymerase 1 Homo sapiens 76-103 20053726-5 2010 Sunitinib treatment also downregulated cyclin E, cyclin D2, and cyclin D3 and upregulated p21Cip1, all of which are involved in regulating cell cycle. Sunitinib 0-9 cyclin D2 Homo sapiens 49-58 20053726-5 2010 Sunitinib treatment also downregulated cyclin E, cyclin D2, and cyclin D3 and upregulated p21Cip1, all of which are involved in regulating cell cycle. Sunitinib 0-9 cyclin D3 Homo sapiens 64-73 20053726-5 2010 Sunitinib treatment also downregulated cyclin E, cyclin D2, and cyclin D3 and upregulated p21Cip1, all of which are involved in regulating cell cycle. Sunitinib 0-9 cyclin dependent kinase inhibitor 1A Homo sapiens 90-97 20053726-7 2010 Dephosphorylation of STAT3 (Tyr(705)) induced by sunitinib was helped by a reduction in activities of Janus-activated kinase 2 and Src. Sunitinib 49-58 signal transducer and activator of transcription 3 Homo sapiens 21-26 20053726-7 2010 Dephosphorylation of STAT3 (Tyr(705)) induced by sunitinib was helped by a reduction in activities of Janus-activated kinase 2 and Src. Sunitinib 49-58 SRC proto-oncogene, non-receptor tyrosine kinase Homo sapiens 131-134 20053726-8 2010 Additionally, sodium vanadate, an inhibitor of protein tyrosine phosphatases, partially blocked the inhibition of phosphorylated STAT3 by sunitinib. Sunitinib 138-147 signal transducer and activator of transcription 3 Homo sapiens 129-134 20053726-9 2010 Loss of phosphorylated AKT after sunitinib treatment was accompanied by decreased phosphorylation of downstream proteins glycogen synthase kinase-3beta and mammalian target of rapamycin. Sunitinib 33-42 AKT serine/threonine kinase 1 Homo sapiens 23-26 20053726-9 2010 Loss of phosphorylated AKT after sunitinib treatment was accompanied by decreased phosphorylation of downstream proteins glycogen synthase kinase-3beta and mammalian target of rapamycin. Sunitinib 33-42 glycogen synthase kinase 3 beta Homo sapiens 121-151 20053726-9 2010 Loss of phosphorylated AKT after sunitinib treatment was accompanied by decreased phosphorylation of downstream proteins glycogen synthase kinase-3beta and mammalian target of rapamycin. Sunitinib 33-42 mechanistic target of rapamycin kinase Homo sapiens 156-185 20215359-3 2010 These include the vascular endothelial growth factor receptor (VEGFR) tyrosine kinase inhibitors sunitinib and sorafenib, which inhibit angiogenic signaling in endothelial cells and vascular pericytes predominantly through VEGFR and platelet-derived growth factor receptor beta. Sunitinib 97-106 kinase insert domain receptor Homo sapiens 63-68 20072832-1 2010 Sunitinib is an oral multikinase inhibitor that blocks the vascular endothelial growth factor receptor (VEGFR), platelet-derived growth factor receptor (PDGFR) alpha and beta, c-kit, and other receptors. Sunitinib 0-9 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 176-181 20173383-0 2010 Plasma N-terminal pro-brain natriuretic peptide as prognostic marker in fatal cardial decompensation with sunitinib malate therapy. Sunitinib 106-122 natriuretic peptide B Homo sapiens 18-47 20072832-1 2010 Sunitinib is an oral multikinase inhibitor that blocks the vascular endothelial growth factor receptor (VEGFR), platelet-derived growth factor receptor (PDGFR) alpha and beta, c-kit, and other receptors. Sunitinib 0-9 kinase insert domain receptor Homo sapiens 59-102 20072832-1 2010 Sunitinib is an oral multikinase inhibitor that blocks the vascular endothelial growth factor receptor (VEGFR), platelet-derived growth factor receptor (PDGFR) alpha and beta, c-kit, and other receptors. Sunitinib 0-9 kinase insert domain receptor Homo sapiens 104-109 20072832-1 2010 Sunitinib is an oral multikinase inhibitor that blocks the vascular endothelial growth factor receptor (VEGFR), platelet-derived growth factor receptor (PDGFR) alpha and beta, c-kit, and other receptors. Sunitinib 0-9 platelet derived growth factor receptor alpha Homo sapiens 153-165 21789125-2 2010 Based on available phase III randomized trials, anti-VEGF agents such as sunitinib, sorafenib, bevacizumab-based therapy, and mTOR-targeted agents such as temsirolimus and everolimus have been used in the treatment armamentarium for this disease. Sunitinib 73-82 vascular endothelial growth factor A Homo sapiens 53-57 20980789-1 2010 INTRODUCTION: We describe our experience with sorafenib and sunitinib in the treatment of chemotherapy-refractory advanced penile squamous cell carcinoma (SCC). Sunitinib 60-69 serpin family B member 3 Homo sapiens 155-158 20980789-10 2010 The patient who achieved partial response had an SCC antigen reduction of nearly 95% after treatment with sunitinib. Sunitinib 106-115 serpin family B member 3 Homo sapiens 49-52 20044640-1 2009 BACKGROUND: The tyrosine kinase inhibitors (TKI) sunitinib and imatinib were shown to induce macrocytosis in patients with renal cell cancer (RCC) and gastrointestinal stromal tumors (GIST), presumably through inhibition of the c-KIT dependent signaling pathway of erythroid progenitor cells of the bone marrow. Sunitinib 49-58 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 228-233 20980789-12 2010 CONCLUSIONS: The feasibility and activity of sorafenib and sunitinib in our series suggest that this approach may be a promising alternative in chemotherapy-refractory advanced penile SCC. Sunitinib 59-68 serpin family B member 3 Homo sapiens 184-187 19911111-1 2009 Sunitinib demonstrating efficacy in pancreatic islet cell carcinomas will pave the way for further trials in other neuroendocrine tumor types such as carcinoid, poorly differentiated neuroendocrine disease, and several other endocrine tumors that are dependent on VEGF/VEGFR for angiogenesis. Sunitinib 0-9 vascular endothelial growth factor A Homo sapiens 264-268 19904265-0 2009 Identification of TNF-alpha and MMP-9 as potential baseline predictive serum markers of sunitinib activity in patients with renal cell carcinoma using a human cytokine array. Sunitinib 88-97 tumor necrosis factor Homo sapiens 18-27 19904265-0 2009 Identification of TNF-alpha and MMP-9 as potential baseline predictive serum markers of sunitinib activity in patients with renal cell carcinoma using a human cytokine array. Sunitinib 88-97 matrix metallopeptidase 9 Homo sapiens 32-37 19904265-9 2009 The area under the ROC curves for TNF-alpha and MMP-9 as predictive markers of sunitinib activity were 0.83 and 0.77. Sunitinib 79-88 tumor necrosis factor Homo sapiens 34-43 19904265-9 2009 The area under the ROC curves for TNF-alpha and MMP-9 as predictive markers of sunitinib activity were 0.83 and 0.77. Sunitinib 79-88 matrix metallopeptidase 9 Homo sapiens 48-53 19954293-7 2009 Multiple new agents targeting the VEGF pathway have been tested and approved, including sunitinib, sorafenib and bevacizumab, with others waiting in the wings. Sunitinib 88-97 vascular endothelial growth factor A Homo sapiens 34-38 19904265-10 2009 CONCLUSION: Baseline levels of TNF-alpha and MMP-9 warrant further study as predictive markers of sunitinib activity in MRCC. Sunitinib 98-107 tumor necrosis factor Homo sapiens 31-40 19904265-10 2009 CONCLUSION: Baseline levels of TNF-alpha and MMP-9 warrant further study as predictive markers of sunitinib activity in MRCC. Sunitinib 98-107 matrix metallopeptidase 9 Homo sapiens 45-50 19911111-1 2009 Sunitinib demonstrating efficacy in pancreatic islet cell carcinomas will pave the way for further trials in other neuroendocrine tumor types such as carcinoid, poorly differentiated neuroendocrine disease, and several other endocrine tumors that are dependent on VEGF/VEGFR for angiogenesis. Sunitinib 0-9 kinase insert domain receptor Homo sapiens 269-274 19767240-0 2009 Risk of bleeding with vascular endothelial growth factor receptor tyrosine-kinase inhibitors sunitinib and sorafenib: a systematic review and meta-analysis of clinical trials. Sunitinib 93-102 kinase insert domain receptor Homo sapiens 22-65 19887629-6 2009 Inhibition of VEGF receptor signaling pathways by sunitinib or VEGFR2 morpholinos virtually completely ablated VEGF-induced tumor cell dissemination and metastasis. Sunitinib 50-59 vascular endothelial growth factor Aa Danio rerio 14-18 19861442-3 2009 The clinical benefit of sunitinib is genotype-dependent in regards to both primary and secondary mutations, with GIST patients harboring the KIT(AY502-3ins) exon 9 mutation being the most sensitive. Sunitinib 24-33 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 141-144 19861442-6 2009 RESULTS: Tumors from patients who developed sunitinib resistance after at least 1 year of radiographic response were analyzed, showing similar findings of a primary KIT(AY502-3ins) mutation and a secondary mutation in the KIT activation loop. Sunitinib 44-53 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 165-168 19861442-6 2009 RESULTS: Tumors from patients who developed sunitinib resistance after at least 1 year of radiographic response were analyzed, showing similar findings of a primary KIT(AY502-3ins) mutation and a secondary mutation in the KIT activation loop. Sunitinib 44-53 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 222-225 19755855-5 2009 Treatment of NCI-H1703 cells with PDGFRA-specific shRNAs or with the PDGFRalpha/KIT small molecule inhibitors imatinib or sunitinib leads to cell growth inhibition. Sunitinib 122-131 platelet derived growth factor receptor alpha Homo sapiens 69-79 19755855-5 2009 Treatment of NCI-H1703 cells with PDGFRA-specific shRNAs or with the PDGFRalpha/KIT small molecule inhibitors imatinib or sunitinib leads to cell growth inhibition. Sunitinib 122-131 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 80-83 19811659-9 2009 The potential targets for other kinase inhibitors (sunitinib and sorafenib) in ChRCC seem to be VEGFR and PDGFR. Sunitinib 51-60 kinase insert domain receptor Homo sapiens 96-101 19811659-9 2009 The potential targets for other kinase inhibitors (sunitinib and sorafenib) in ChRCC seem to be VEGFR and PDGFR. Sunitinib 51-60 platelet derived growth factor receptor beta Homo sapiens 106-111 19844230-6 2009 RESULTS: Sunitinib inhibited more kinases than pazopanib and sorafenib, at potencies within 10-fold of VEGFR-2. Sunitinib 9-18 kinase insert domain receptor Homo sapiens 103-110 19844230-9 2009 Addition of stem cell factor and/or Flt-3 ligand with granulocyte-macrophage colony stimulating factor resulted in significant shifts in potency for sorafenib and sunitinib but less so for pazopanib. Sunitinib 163-172 KIT ligand Homo sapiens 12-28 19844230-9 2009 Addition of stem cell factor and/or Flt-3 ligand with granulocyte-macrophage colony stimulating factor resulted in significant shifts in potency for sorafenib and sunitinib but less so for pazopanib. Sunitinib 163-172 fms related receptor tyrosine kinase 3 Homo sapiens 36-41 19773375-1 2009 PURPOSE: Bevacizumab is an antibody against vascular endothelial growth factor; sunitinib is an inhibitor of vascular endothelial growth factor and related receptors. Sunitinib 80-89 vascular endothelial growth factor A Homo sapiens 109-143 19773380-6 2009 Sorafenib and sunitinib showed concentration-dependent (1 and 10 micromol/L), low to moderate affinity for ABCB1 but were not affected by the other ABC transporters. Sunitinib 14-23 ATP binding cassette subfamily B member 1 Homo sapiens 107-112 19767240-1 2009 BACKGROUND: Sunitinib and sorafenib are oral vascular endothelial growth factor receptor (VEGFR) tyrosine-kinase inhibitors used in various cancers. Sunitinib 12-21 kinase insert domain receptor Homo sapiens 45-88 19767240-1 2009 BACKGROUND: Sunitinib and sorafenib are oral vascular endothelial growth factor receptor (VEGFR) tyrosine-kinase inhibitors used in various cancers. Sunitinib 12-21 kinase insert domain receptor Homo sapiens 90-95 19767240-11 2009 INTERPRETATION: Treatment with the VEGFR tyrosine-kinase inhibitors sunitinib and sorafenib is associated with a significant increase in risk of bleeding. Sunitinib 68-77 kinase insert domain receptor Homo sapiens 35-40 19481151-1 2009 Several antiangiogenic agents, including bevacizumab, sunitinib, and sorafenib, which mainly target the VEGF signaling system, have been approved for the treatment of human cancers. Sunitinib 54-63 vascular endothelial growth factor A Homo sapiens 104-108 19774211-2 2009 This breakthrough in science led to the development of a variety of small molecules inhibiting the VEGF-dependent angiogenic pathway, such as sunitinib and sorafenib. Sunitinib 142-151 vascular endothelial growth factor A Homo sapiens 99-103 19754358-2 2009 Since HCC is a particularly vascular solid tumor, we determined the antitumor and antiangiogenic activities of sunitinib malate, a potent inhibitor of two receptors involved in angiogenesis - vascular endothelial growth factor receptor (VEGFR) and platelet-derived growth factor receptor (PDGFR). Sunitinib 111-127 kinase insert domain receptor Homo sapiens 192-235 19737946-7 2009 Sunitinib treatment was associated with reduced tumor cell proliferation by >25% in 52% of cases analyzed and reduced levels of phospho-KIT in tumor biopsies (indicating target modulation). Sunitinib 0-9 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 139-142 19737946-11 2009 CONCLUSION: Cellular and molecular analyses showed that sunitinib clinical activity is associated with inhibition of KIT in GIST following imatinib failure, illustrating the rational approach used to develop a therapy aimed at the underlying oncogenic signaling pathway aberrancy. Sunitinib 56-65 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 117-120 19737953-0 2009 Circulating levels of soluble KIT serve as a biomarker for clinical outcome in gastrointestinal stromal tumor patients receiving sunitinib following imatinib failure. Sunitinib 129-138 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 30-33 19737953-1 2009 PURPOSE: To evaluate changes in circulating levels of soluble KIT (sKIT) extracellular domain as a potential biomarker for clinical outcome in gastrointestinal stromal tumor patients treated with the multitargeted tyrosine kinase inhibitor sunitinib following imatinib failure in a previously reported phase III study. Sunitinib 240-249 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 62-65 19754358-2 2009 Since HCC is a particularly vascular solid tumor, we determined the antitumor and antiangiogenic activities of sunitinib malate, a potent inhibitor of two receptors involved in angiogenesis - vascular endothelial growth factor receptor (VEGFR) and platelet-derived growth factor receptor (PDGFR). Sunitinib 111-127 kinase insert domain receptor Homo sapiens 237-242 19754358-2 2009 Since HCC is a particularly vascular solid tumor, we determined the antitumor and antiangiogenic activities of sunitinib malate, a potent inhibitor of two receptors involved in angiogenesis - vascular endothelial growth factor receptor (VEGFR) and platelet-derived growth factor receptor (PDGFR). Sunitinib 111-127 platelet derived growth factor receptor beta Homo sapiens 248-287 19754358-2 2009 Since HCC is a particularly vascular solid tumor, we determined the antitumor and antiangiogenic activities of sunitinib malate, a potent inhibitor of two receptors involved in angiogenesis - vascular endothelial growth factor receptor (VEGFR) and platelet-derived growth factor receptor (PDGFR). Sunitinib 111-127 platelet derived growth factor receptor beta Homo sapiens 289-294 19754358-4 2009 Sunitinib inhibited phosphorylation of VEGFR-2 at Tyr951 and PDGFR-beta at Tyr1021 both in vitro and in vivo. Sunitinib 0-9 kinase insert domain receptor Homo sapiens 39-46 19754358-4 2009 Sunitinib inhibited phosphorylation of VEGFR-2 at Tyr951 and PDGFR-beta at Tyr1021 both in vitro and in vivo. Sunitinib 0-9 platelet derived growth factor receptor beta Homo sapiens 61-71 19724685-9 2009 KCI-18 cells and tumors expressed vascular endothelial growth factor receptor 2 and platelet-derived growth factor receptor beta molecular targets of sunitinib that were modulated by the drug treatment. Sunitinib 150-159 kinase insert domain protein receptor Mus musculus 34-128 19672141-2 2009 Sunitinib malate is an oral, multitargeted inhibitor of vascular endothelial growth factor receptor-1, -2, and -3, platelet-derived growth factor receptor-alpha and -beta, and other kinases implicated in tumor growth, angiogenesis, and metastasis. Sunitinib 0-16 platelet derived growth factor receptor alpha Homo sapiens 115-170 19726307-3 2009 The inhibitory activity of Sunitinib, a standard PTK inhibitor, on vascular endothelia growth factor receptor 2 (VEGFR-2) kinase activity was investigated. Sunitinib 27-36 kinase insert domain receptor Homo sapiens 67-111 19726307-3 2009 The inhibitory activity of Sunitinib, a standard PTK inhibitor, on vascular endothelia growth factor receptor 2 (VEGFR-2) kinase activity was investigated. Sunitinib 27-36 kinase insert domain receptor Homo sapiens 113-120 19726307-6 2009 Sunitinib inhibited VEGFR-2 kinase activity with an IC50 value of 86.7 nmol/L, which was close to the values tested using other methods. Sunitinib 0-9 kinase insert domain receptor Homo sapiens 20-27 19672141-2 2009 Sunitinib malate is an oral, multitargeted inhibitor of vascular endothelial growth factor receptor-1, -2, and -3, platelet-derived growth factor receptor-alpha and -beta, and other kinases implicated in tumor growth, angiogenesis, and metastasis. Sunitinib 0-16 fms related receptor tyrosine kinase 1 Homo sapiens 56-113 19617296-5 2009 Receptor tyrosine-kinase inhibitors (imatinib, dasatinib, sunitinib) may have a role in treatment of patients with melanoma harbouring c-Kit mutations. Sunitinib 58-67 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 135-140 19568242-14 2009 CONCLUSION: There is evidence to suggest that treatment with sunitinib and treatment with bevacizumab plus IFN has clinically relevant and statistically significant advantages over treatment with IFN alone in patients with metastatic RCC. Sunitinib 61-70 interferon alpha 1 Homo sapiens 196-199 19689244-6 2009 In contrast, folliculitis is not common during administration of sorafenib and sunitinib, which target VEGFR, PDGFR, FLT3, and others, whereas both agents have been associated with subungual splinter hemorrhages. Sunitinib 79-88 kinase insert domain receptor Homo sapiens 103-108 19232821-0 2009 Sensitization of ABCG2-overexpressing cells to conventional chemotherapeutic agent by sunitinib was associated with inhibiting the function of ABCG2. Sunitinib 86-95 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 17-22 19326424-1 2009 BACKGROUND: Sunitinib inhibits KIT and other members of the split-kinase-domain family of receptor tyrosine kinases. Sunitinib 12-21 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 31-34 21789110-3 2009 Bevacizumab is directed against the vascular endothelial growth factor (VEGF), a key mediator of angiogenesis, whilst sorafenib and sunitinib inhibit a number of targets including the VEGF and platelet-derived growth factor (PDGFR) receptor tyrosine kinases. Sunitinib 132-141 vascular endothelial growth factor A Homo sapiens 184-188 21789110-3 2009 Bevacizumab is directed against the vascular endothelial growth factor (VEGF), a key mediator of angiogenesis, whilst sorafenib and sunitinib inhibit a number of targets including the VEGF and platelet-derived growth factor (PDGFR) receptor tyrosine kinases. Sunitinib 132-141 platelet derived growth factor receptor beta Homo sapiens 193-223 18534874-1 2009 PURPOSE: Sunitinib malate (Pfizer, Inc.) is a multitargeted kinase inhibitor that inhibits vascular endothelial growth factor (VEGF) receptor (R)-1, 2 and 3, platelet-derived growth factor receptors (PDGFR)-alpha and beta, Flt3, RET, and Kit. Sunitinib 9-25 vascular endothelial growth factor A Homo sapiens 127-131 18534874-1 2009 PURPOSE: Sunitinib malate (Pfizer, Inc.) is a multitargeted kinase inhibitor that inhibits vascular endothelial growth factor (VEGF) receptor (R)-1, 2 and 3, platelet-derived growth factor receptors (PDGFR)-alpha and beta, Flt3, RET, and Kit. Sunitinib 9-25 platelet derived growth factor receptor alpha Homo sapiens 200-212 18534874-1 2009 PURPOSE: Sunitinib malate (Pfizer, Inc.) is a multitargeted kinase inhibitor that inhibits vascular endothelial growth factor (VEGF) receptor (R)-1, 2 and 3, platelet-derived growth factor receptors (PDGFR)-alpha and beta, Flt3, RET, and Kit. Sunitinib 9-25 fms related receptor tyrosine kinase 3 Homo sapiens 223-227 18534874-1 2009 PURPOSE: Sunitinib malate (Pfizer, Inc.) is a multitargeted kinase inhibitor that inhibits vascular endothelial growth factor (VEGF) receptor (R)-1, 2 and 3, platelet-derived growth factor receptors (PDGFR)-alpha and beta, Flt3, RET, and Kit. Sunitinib 9-25 ret proto-oncogene Homo sapiens 229-232 19232821-0 2009 Sensitization of ABCG2-overexpressing cells to conventional chemotherapeutic agent by sunitinib was associated with inhibiting the function of ABCG2. Sunitinib 86-95 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 143-148 19232821-2 2009 In this study, we evaluated the possible interaction of sunitinib with P-glycoprotein (P-gp, ABCB1), multidrug resistance protein 1 (MRP1, ABCC1), breast cancer resistance protein (BCRP, ABCG2) and lung-resistance protein (LRP) in vitro. Sunitinib 56-65 ATP binding cassette subfamily B member 1 Homo sapiens 71-85 19232821-2 2009 In this study, we evaluated the possible interaction of sunitinib with P-glycoprotein (P-gp, ABCB1), multidrug resistance protein 1 (MRP1, ABCC1), breast cancer resistance protein (BCRP, ABCG2) and lung-resistance protein (LRP) in vitro. Sunitinib 56-65 ATP binding cassette subfamily B member 1 Homo sapiens 87-91 19232821-2 2009 In this study, we evaluated the possible interaction of sunitinib with P-glycoprotein (P-gp, ABCB1), multidrug resistance protein 1 (MRP1, ABCC1), breast cancer resistance protein (BCRP, ABCG2) and lung-resistance protein (LRP) in vitro. Sunitinib 56-65 ATP binding cassette subfamily B member 1 Homo sapiens 93-98 19232821-2 2009 In this study, we evaluated the possible interaction of sunitinib with P-glycoprotein (P-gp, ABCB1), multidrug resistance protein 1 (MRP1, ABCC1), breast cancer resistance protein (BCRP, ABCG2) and lung-resistance protein (LRP) in vitro. Sunitinib 56-65 ATP binding cassette subfamily B member 1 Homo sapiens 101-131 19232821-2 2009 In this study, we evaluated the possible interaction of sunitinib with P-glycoprotein (P-gp, ABCB1), multidrug resistance protein 1 (MRP1, ABCC1), breast cancer resistance protein (BCRP, ABCG2) and lung-resistance protein (LRP) in vitro. Sunitinib 56-65 ATP binding cassette subfamily B member 1 Homo sapiens 133-137 19232821-2 2009 In this study, we evaluated the possible interaction of sunitinib with P-glycoprotein (P-gp, ABCB1), multidrug resistance protein 1 (MRP1, ABCC1), breast cancer resistance protein (BCRP, ABCG2) and lung-resistance protein (LRP) in vitro. Sunitinib 56-65 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 181-185 19232821-2 2009 In this study, we evaluated the possible interaction of sunitinib with P-glycoprotein (P-gp, ABCB1), multidrug resistance protein 1 (MRP1, ABCC1), breast cancer resistance protein (BCRP, ABCG2) and lung-resistance protein (LRP) in vitro. Sunitinib 56-65 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 187-192 19232821-2 2009 In this study, we evaluated the possible interaction of sunitinib with P-glycoprotein (P-gp, ABCB1), multidrug resistance protein 1 (MRP1, ABCC1), breast cancer resistance protein (BCRP, ABCG2) and lung-resistance protein (LRP) in vitro. Sunitinib 56-65 major vault protein Homo sapiens 198-221 19232821-2 2009 In this study, we evaluated the possible interaction of sunitinib with P-glycoprotein (P-gp, ABCB1), multidrug resistance protein 1 (MRP1, ABCC1), breast cancer resistance protein (BCRP, ABCG2) and lung-resistance protein (LRP) in vitro. Sunitinib 56-65 major vault protein Homo sapiens 223-226 19232821-3 2009 Our results showed that sunitinib completely reverse drug resistance mediated by ABCG2 at a non-toxic concentration of 2.5muM and has no significant reversal effect on ABCB1-, ABCC1- and LRP-mediated drug resistance, although a small synergetic effect was observed in combining sunitinib and conventional chemotherapeutic agents in ABCB1 overexpressing MCF-7/adr and parental sensitive MCF-7 cells, ABCC1 overexpressing C-A120 and parental sensitive KB-3-1 cells. Sunitinib 24-33 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 81-86 19232821-4 2009 Sunitinib significantly increased intracellular accumulation of rhodamine 123 and doxorubicin and remarkably inhibited the efflux of rhodamine 123 and methotrexate by ABCG2 in ABCG2-overexpressing cells, and also profoundly inhibited the transport of [(3)H]-methotrexate by ABCG2. Sunitinib 0-9 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 167-172 19232821-4 2009 Sunitinib significantly increased intracellular accumulation of rhodamine 123 and doxorubicin and remarkably inhibited the efflux of rhodamine 123 and methotrexate by ABCG2 in ABCG2-overexpressing cells, and also profoundly inhibited the transport of [(3)H]-methotrexate by ABCG2. Sunitinib 0-9 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 176-181 19232821-4 2009 Sunitinib significantly increased intracellular accumulation of rhodamine 123 and doxorubicin and remarkably inhibited the efflux of rhodamine 123 and methotrexate by ABCG2 in ABCG2-overexpressing cells, and also profoundly inhibited the transport of [(3)H]-methotrexate by ABCG2. Sunitinib 0-9 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 176-181 19232821-7 2009 Overall, we conclude that sunitinib reverses ABCG2-mediated MDR through inhibiting the drug efflux function of ABCG2. Sunitinib 26-35 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 45-50 19232821-7 2009 Overall, we conclude that sunitinib reverses ABCG2-mediated MDR through inhibiting the drug efflux function of ABCG2. Sunitinib 26-35 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 111-116 19453268-3 2009 Sunitinib is a multi-kinase inhibitor of VEGFR-2, PDGFR (alpha,beta), FLT-3, KIT, CSF-1 and RET. Sunitinib 0-9 kinase insert domain receptor Homo sapiens 41-48 19467916-5 2009 We discuss how the cross-talk between major downstream signaling pathways, such as PI3K/PTEN/Akt/mTOR, RAS/Raf/MEK/ERK, and Jak/STAT, can be exploited for therapeutic purposes by targeting key signaling molecules with selective inhibitors, such as mTOR inhibitors, HSP90 inhibitors, or farnesyltransferase inhibitors, and identifying those agents with the ability to positively combine with inhibitors of FLT3, such as PKC412 and sunitinib. Sunitinib 430-439 phosphatase and tensin homolog Homo sapiens 88-92 19467916-5 2009 We discuss how the cross-talk between major downstream signaling pathways, such as PI3K/PTEN/Akt/mTOR, RAS/Raf/MEK/ERK, and Jak/STAT, can be exploited for therapeutic purposes by targeting key signaling molecules with selective inhibitors, such as mTOR inhibitors, HSP90 inhibitors, or farnesyltransferase inhibitors, and identifying those agents with the ability to positively combine with inhibitors of FLT3, such as PKC412 and sunitinib. Sunitinib 430-439 AKT serine/threonine kinase 1 Homo sapiens 93-96 19467916-5 2009 We discuss how the cross-talk between major downstream signaling pathways, such as PI3K/PTEN/Akt/mTOR, RAS/Raf/MEK/ERK, and Jak/STAT, can be exploited for therapeutic purposes by targeting key signaling molecules with selective inhibitors, such as mTOR inhibitors, HSP90 inhibitors, or farnesyltransferase inhibitors, and identifying those agents with the ability to positively combine with inhibitors of FLT3, such as PKC412 and sunitinib. Sunitinib 430-439 mechanistic target of rapamycin kinase Homo sapiens 97-101 19467916-5 2009 We discuss how the cross-talk between major downstream signaling pathways, such as PI3K/PTEN/Akt/mTOR, RAS/Raf/MEK/ERK, and Jak/STAT, can be exploited for therapeutic purposes by targeting key signaling molecules with selective inhibitors, such as mTOR inhibitors, HSP90 inhibitors, or farnesyltransferase inhibitors, and identifying those agents with the ability to positively combine with inhibitors of FLT3, such as PKC412 and sunitinib. Sunitinib 430-439 zinc fingers and homeoboxes 2 Homo sapiens 107-110 19467916-5 2009 We discuss how the cross-talk between major downstream signaling pathways, such as PI3K/PTEN/Akt/mTOR, RAS/Raf/MEK/ERK, and Jak/STAT, can be exploited for therapeutic purposes by targeting key signaling molecules with selective inhibitors, such as mTOR inhibitors, HSP90 inhibitors, or farnesyltransferase inhibitors, and identifying those agents with the ability to positively combine with inhibitors of FLT3, such as PKC412 and sunitinib. Sunitinib 430-439 mitogen-activated protein kinase kinase 7 Homo sapiens 111-114 19467916-5 2009 We discuss how the cross-talk between major downstream signaling pathways, such as PI3K/PTEN/Akt/mTOR, RAS/Raf/MEK/ERK, and Jak/STAT, can be exploited for therapeutic purposes by targeting key signaling molecules with selective inhibitors, such as mTOR inhibitors, HSP90 inhibitors, or farnesyltransferase inhibitors, and identifying those agents with the ability to positively combine with inhibitors of FLT3, such as PKC412 and sunitinib. Sunitinib 430-439 EPH receptor B2 Homo sapiens 115-118 19467916-5 2009 We discuss how the cross-talk between major downstream signaling pathways, such as PI3K/PTEN/Akt/mTOR, RAS/Raf/MEK/ERK, and Jak/STAT, can be exploited for therapeutic purposes by targeting key signaling molecules with selective inhibitors, such as mTOR inhibitors, HSP90 inhibitors, or farnesyltransferase inhibitors, and identifying those agents with the ability to positively combine with inhibitors of FLT3, such as PKC412 and sunitinib. Sunitinib 430-439 mechanistic target of rapamycin kinase Homo sapiens 248-252 19467916-5 2009 We discuss how the cross-talk between major downstream signaling pathways, such as PI3K/PTEN/Akt/mTOR, RAS/Raf/MEK/ERK, and Jak/STAT, can be exploited for therapeutic purposes by targeting key signaling molecules with selective inhibitors, such as mTOR inhibitors, HSP90 inhibitors, or farnesyltransferase inhibitors, and identifying those agents with the ability to positively combine with inhibitors of FLT3, such as PKC412 and sunitinib. Sunitinib 430-439 heat shock protein 90 alpha family class A member 1 Homo sapiens 265-270 19467916-5 2009 We discuss how the cross-talk between major downstream signaling pathways, such as PI3K/PTEN/Akt/mTOR, RAS/Raf/MEK/ERK, and Jak/STAT, can be exploited for therapeutic purposes by targeting key signaling molecules with selective inhibitors, such as mTOR inhibitors, HSP90 inhibitors, or farnesyltransferase inhibitors, and identifying those agents with the ability to positively combine with inhibitors of FLT3, such as PKC412 and sunitinib. Sunitinib 430-439 fms related receptor tyrosine kinase 3 Homo sapiens 405-409 19496707-5 2009 It has been tested in patients with progressive disease after therapy with tyrosine kinase receptor inhibitors (sunitinib, sorafenib or both), which interfere with signaling pathways, such as the VEGF pathway. Sunitinib 112-121 vascular endothelial growth factor A Homo sapiens 196-200 19318229-3 2009 GI50 values were determined: ZD6474 and SU11248, mainly VEGFR2 inhibitors, gave the lowest GI50 across all cell lines (3.5-6.9 microM) whereas ZD1839 gave a GI50 in this range only in H28 cells. Sunitinib 40-47 kinase insert domain receptor Homo sapiens 56-62 19318229-5 2009 ZD6474 and ZD1839 inhibited EGF-induced phosphorylation of EGFR, AKT and ERK, whereas VEGF-induced phosphorylation of VEGFR2 was completely inhibited with 0.1 microM SU11248. Sunitinib 166-173 vascular endothelial growth factor A Homo sapiens 86-90 19318229-5 2009 ZD6474 and ZD1839 inhibited EGF-induced phosphorylation of EGFR, AKT and ERK, whereas VEGF-induced phosphorylation of VEGFR2 was completely inhibited with 0.1 microM SU11248. Sunitinib 166-173 kinase insert domain receptor Homo sapiens 118-124 19453268-3 2009 Sunitinib is a multi-kinase inhibitor of VEGFR-2, PDGFR (alpha,beta), FLT-3, KIT, CSF-1 and RET. Sunitinib 0-9 platelet derived growth factor receptor beta Homo sapiens 50-55 19453268-3 2009 Sunitinib is a multi-kinase inhibitor of VEGFR-2, PDGFR (alpha,beta), FLT-3, KIT, CSF-1 and RET. Sunitinib 0-9 fms related receptor tyrosine kinase 3 Homo sapiens 70-75 19453268-3 2009 Sunitinib is a multi-kinase inhibitor of VEGFR-2, PDGFR (alpha,beta), FLT-3, KIT, CSF-1 and RET. Sunitinib 0-9 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 77-80 19453268-3 2009 Sunitinib is a multi-kinase inhibitor of VEGFR-2, PDGFR (alpha,beta), FLT-3, KIT, CSF-1 and RET. Sunitinib 0-9 colony stimulating factor 1 Homo sapiens 82-87 19453268-3 2009 Sunitinib is a multi-kinase inhibitor of VEGFR-2, PDGFR (alpha,beta), FLT-3, KIT, CSF-1 and RET. Sunitinib 0-9 ret proto-oncogene Homo sapiens 92-95 19171421-4 2009 Both Sorafenib and Sunitinib with Enzastaurin at concentrations feasible in vivo showed a synergistic reduction of viable RCC cells by inhibiting cell growth through inhibition of phospho-S6-kinase and GSK3-beta. Sunitinib 19-28 glycogen synthase kinase 3 beta Homo sapiens 202-211 19493441-0 2009 Inhibitory effects of sunitinib on ovalbumin-induced chronic experimental asthma in mice. Sunitinib 22-31 serine (or cysteine) peptidase inhibitor, clade B, member 1, pseudogene Mus musculus 35-44 19402059-2 2009 Sunitinib malate, sorafenib tosylate, bevacizumab with interferon alpha, temsirolimus, and everolimus have improved clinical outcomes in randomized phase 3 trials by inhibiting the vascular endothelial growth factor and related pathways. Sunitinib 0-16 vascular endothelial growth factor A Homo sapiens 181-215 19402073-3 2009 Approaches to bind circulating VEGF protein (eg, bevacizumab) and small molecule inhibitors of the receptor on which the VEGF ligand binds (eg, sunitinib, sorafenib, axitinib, and pazopanib) have been tested. Sunitinib 144-153 vascular endothelial growth factor A Homo sapiens 121-125 19403935-10 2009 Significant changes following sunitinib treatment were observed in serum-soluble biomarkers including soluble vascular endothelial growth factor receptor-2, platelet-derived growth factor aa, placental growth factor and leptin. Sunitinib 30-39 placental growth factor Homo sapiens 192-215 19366796-2 2009 Sunitinib is a clinically approved multitargeted tyrosine kinase inhibitor that inhibits vascular endothelial growth factor receptor, c-KIT, and PDGFR, and has shown clinical activity in various solid tumors. Sunitinib 0-9 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 134-139 19366796-2 2009 Sunitinib is a clinically approved multitargeted tyrosine kinase inhibitor that inhibits vascular endothelial growth factor receptor, c-KIT, and PDGFR, and has shown clinical activity in various solid tumors. Sunitinib 0-9 platelet derived growth factor receptor beta Homo sapiens 145-150 19366796-3 2009 Activation of PDGFR signaling has been described in gastrointestinal stromal tumors (PDGFRA mutations) as well as in chronic myeloid leukemia (BCR-PDGFRA translocation), and sunitinib can yield clinical benefit in both settings. Sunitinib 174-183 platelet derived growth factor receptor beta Homo sapiens 14-19 19366796-4 2009 However, the discovery of PDGFR activating mutations or gene rearrangements in other tumor types could reveal additional patient populations who might benefit from treatment with anti-PDGFR therapies, such as sunitinib. Sunitinib 209-218 platelet derived growth factor receptor beta Homo sapiens 26-31 19458500-0 2009 Sunitinib and PF-562,271 (FAK/Pyk2 inhibitor) effectively block growth and recovery of human hepatocellular carcinoma in a rat xenograft model. Sunitinib 0-9 protein tyrosine kinase 2 Homo sapiens 26-29 19366796-4 2009 However, the discovery of PDGFR activating mutations or gene rearrangements in other tumor types could reveal additional patient populations who might benefit from treatment with anti-PDGFR therapies, such as sunitinib. Sunitinib 209-218 platelet derived growth factor receptor beta Homo sapiens 184-189 19458500-0 2009 Sunitinib and PF-562,271 (FAK/Pyk2 inhibitor) effectively block growth and recovery of human hepatocellular carcinoma in a rat xenograft model. Sunitinib 0-9 protein tyrosine kinase 2 beta Homo sapiens 30-34 19366796-7 2009 In the sunitinib-sensitive adenosquamous NSCLC cell line, PDGFRA expression was associated with focal PFGRA gene amplification, which was similarly detected in a small fraction of squamous cell NSCLC primary tumor specimens. Sunitinib 7-16 platelet derived growth factor receptor alpha Homo sapiens 58-64 19366796-9 2009 A similar codependency on PDGFRA and PDGFC was observed in the sunitinib-sensitive rhabdomyosarcoma cell line. Sunitinib 63-72 platelet derived growth factor receptor alpha Homo sapiens 26-32 19366796-9 2009 A similar codependency on PDGFRA and PDGFC was observed in the sunitinib-sensitive rhabdomyosarcoma cell line. Sunitinib 63-72 platelet derived growth factor C Homo sapiens 37-42 19435875-7 2009 We found that disruption of HIF-1alpha, HIF-2alpha, or both HIF-1alpha and HIF-2alpha genes led to improved tumor response to sunitinib. Sunitinib 126-135 hypoxia inducible factor 1 subunit alpha Homo sapiens 28-38 19336014-2 2009 Dissection of the molecular pathways that regulate proliferation, apoptosis, and angiogenesis has led to the development of targeted therapies such as the receptor tyrosine kinase inhibitors sunitinib and sorafenib, the anti-vascular endothelial growth factor antibody bevacizumab, and a class of rapamycin analogues including everolimus and temsirolimus. Sunitinib 191-200 vascular endothelial growth factor A Homo sapiens 225-259 19435875-7 2009 We found that disruption of HIF-1alpha, HIF-2alpha, or both HIF-1alpha and HIF-2alpha genes led to improved tumor response to sunitinib. Sunitinib 126-135 endothelial PAS domain protein 1 Homo sapiens 40-50 19435875-8 2009 For xenografts in which both HIF-1alpha and HIF-2alpha genes were disrupted, there was prolonged complete remission with sunitinib treatment in 50% of mice. Sunitinib 121-130 hypoxia inducible factor 1, alpha subunit Mus musculus 29-39 19435875-8 2009 For xenografts in which both HIF-1alpha and HIF-2alpha genes were disrupted, there was prolonged complete remission with sunitinib treatment in 50% of mice. Sunitinib 121-130 endothelial PAS domain protein 1 Mus musculus 44-54 19435875-10 2009 First, tumor angiogenesis and perfusion were almost completely inhibited by sunitinib when both HIF-1alpha and HIF-2alpha genes were disrupted. Sunitinib 76-85 hypoxia inducible factor 1 subunit alpha Homo sapiens 96-106 19435875-10 2009 First, tumor angiogenesis and perfusion were almost completely inhibited by sunitinib when both HIF-1alpha and HIF-2alpha genes were disrupted. Sunitinib 76-85 endothelial PAS domain protein 1 Homo sapiens 111-121 19277038-2 2009 Bevacizumab, sorafenib and sunitinib target VEGF-mediated angiogenesis and are active against several types of cancer, but their effects on the immune system are poorly understood. Sunitinib 27-36 vascular endothelial growth factor A Homo sapiens 44-48 19412427-11 2009 Low-dose sunitinib (20 mg/kg) demonstrates synergistic cytotoxicity with an mTOR inhibitor, rapamycin, which is more effective than the traditional chemotherapeutic drug, cyclophosphamide. Sunitinib 9-18 mechanistic target of rapamycin kinase Homo sapiens 76-80 19336729-5 2009 We showed that daily treatment with sunitinib reduced tumor size, caused tumor necrosis, blocked tumor progression, and prolonged median survival in both the metastatic (Lkb1/Kras) and nonmetastatic (Kras) mouse models; median survival was not reached in the nonmetastatic model after 1 year. Sunitinib 36-45 serine/threonine kinase 11 Mus musculus 170-174 19318574-9 2009 TK1 mutations exhibited a differential response to SU5614, sorafenib, and sunitinib but strongly impaired response to PKC412. Sunitinib 74-83 thymidine kinase 1 Homo sapiens 0-3 19336729-5 2009 We showed that daily treatment with sunitinib reduced tumor size, caused tumor necrosis, blocked tumor progression, and prolonged median survival in both the metastatic (Lkb1/Kras) and nonmetastatic (Kras) mouse models; median survival was not reached in the nonmetastatic model after 1 year. Sunitinib 36-45 Kirsten rat sarcoma viral oncogene homolog Mus musculus 175-179 19318574-10 2009 TK2 exchanges identified with SU5614 were sensitive to PKC412, sunitinib, or sorafenib, with the exception of Y842D, which caused a strong resistance to sorafenib. Sunitinib 63-72 thymidine kinase 2 Homo sapiens 0-3 19336729-5 2009 We showed that daily treatment with sunitinib reduced tumor size, caused tumor necrosis, blocked tumor progression, and prolonged median survival in both the metastatic (Lkb1/Kras) and nonmetastatic (Kras) mouse models; median survival was not reached in the nonmetastatic model after 1 year. Sunitinib 36-45 Kirsten rat sarcoma viral oncogene homolog Mus musculus 200-204 19335275-4 2009 In this situation the second-line TKI sunitinib is well suited to patients with KIT exon 9 mutations, or for patients without KIT/PDGFRA mutations (wild-type GIST). Sunitinib 38-47 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 80-83 19258444-2 2009 Following administration, sunitinib is metabolized by cytochrome P450 3A4 to an active metabolite (SU12662). Sunitinib 26-35 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 54-73 19335275-4 2009 In this situation the second-line TKI sunitinib is well suited to patients with KIT exon 9 mutations, or for patients without KIT/PDGFRA mutations (wild-type GIST). Sunitinib 38-47 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 126-129 19335275-4 2009 In this situation the second-line TKI sunitinib is well suited to patients with KIT exon 9 mutations, or for patients without KIT/PDGFRA mutations (wild-type GIST). Sunitinib 38-47 platelet derived growth factor receptor alpha Homo sapiens 130-136 19224847-1 2009 PURPOSE: Both bevacizumab and sunitinib target the vascular endothelial growth factor pathway and demonstrate activity against advanced renal cell carcinoma (RCC). Sunitinib 30-39 vascular endothelial growth factor A Homo sapiens 51-85 19381023-3 2009 However, cis-mutations in the activation loop of the KIT gene tend to develop Sunitinib-resistant GIST. Sunitinib 78-87 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 53-56 19390946-0 2009 Sunitinib-resistant gastrointestinal stromal tumors harbor cis-mutations in the activation loop of the KIT gene. Sunitinib 0-9 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 103-106 19390946-4 2009 RESULTS: All imatinib- as well as sunitinib-resistant lesions showed viable tumor cells strongly re-expressing the KIT protein. Sunitinib 34-43 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 115-118 19390946-7 2009 A tumor with mutations in exon 11 and 13 of the KIT gene, and showing partial response to sunitinib, harbored a third mutation in the activation loop when sunitinib resistance was shown. Sunitinib 90-99 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 48-51 19390946-7 2009 A tumor with mutations in exon 11 and 13 of the KIT gene, and showing partial response to sunitinib, harbored a third mutation in the activation loop when sunitinib resistance was shown. Sunitinib 155-164 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 48-51 19390946-9 2009 CONCLUSION: These findings indicate that an additional cis-mutation in the activation loop of the KIT gene could be a potential cause of sunitinib resistance in gastrointestinal stromal tumors. Sunitinib 137-146 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 98-101 19244102-0 2009 Sunitinib inhibition of Stat3 induces renal cell carcinoma tumor cell apoptosis and reduces immunosuppressive cells. Sunitinib 0-9 signal transducer and activator of transcription 3 Mus musculus 24-29 19244102-4 2009 We show that sunitinib induces tumor cell apoptosis and growth arrest in RCC tumor cells, which correlates with signal transducer and activator of transcription 3 (Stat3) activity inhibition. Sunitinib 13-22 signal transducer and activator of transcription 3 Mus musculus 112-162 19244102-4 2009 We show that sunitinib induces tumor cell apoptosis and growth arrest in RCC tumor cells, which correlates with signal transducer and activator of transcription 3 (Stat3) activity inhibition. Sunitinib 13-22 signal transducer and activator of transcription 3 Mus musculus 164-169 19244102-6 2009 Reduction of Stat3 activity enhances the antitumor effects of sunitinib, whereas expression of a constitutively activated Stat3 mutant rescues tumor cell death. Sunitinib 62-71 signal transducer and activator of transcription 3 Mus musculus 13-18 19276342-6 2009 Interestingly, sunitinib treatment resulted in reduced expression of interleukin (IL)-10, transforming growth factor-beta, and Foxp3 but enhanced expression of Th1 cytokine IFN-gamma and increased CTL responses in isolated tumor-infiltrating leukocytes. Sunitinib 15-24 interleukin 10 Mus musculus 69-88 19244102-8 2009 Sunitinib also inhibits Stat3 in Renca tumor-associated myeloid-derived suppressor cells (MDSC), down-regulates angiogenic gene expression, and reduces MDSCs and tumor T regulatory cells. Sunitinib 0-9 signal transducer and activator of transcription 3 Mus musculus 24-29 19276342-6 2009 Interestingly, sunitinib treatment resulted in reduced expression of interleukin (IL)-10, transforming growth factor-beta, and Foxp3 but enhanced expression of Th1 cytokine IFN-gamma and increased CTL responses in isolated tumor-infiltrating leukocytes. Sunitinib 15-24 forkhead box P3 Mus musculus 127-132 19276342-6 2009 Interestingly, sunitinib treatment resulted in reduced expression of interleukin (IL)-10, transforming growth factor-beta, and Foxp3 but enhanced expression of Th1 cytokine IFN-gamma and increased CTL responses in isolated tumor-infiltrating leukocytes. Sunitinib 15-24 negative elongation factor complex member C/D, Th1l Mus musculus 160-163 19276342-6 2009 Interestingly, sunitinib treatment resulted in reduced expression of interleukin (IL)-10, transforming growth factor-beta, and Foxp3 but enhanced expression of Th1 cytokine IFN-gamma and increased CTL responses in isolated tumor-infiltrating leukocytes. Sunitinib 15-24 interferon gamma Mus musculus 173-182 19276342-7 2009 A significantly higher percentage and infiltration of CD8 and CD4 cells was detected in tumors of sunitinib-treated mice when compared with control-treated mice. Sunitinib 98-107 CD4 molecule Sus scrofa 62-65 19244102-9 2009 These results suggest that Stat3 activity is important for RCC response to sunitinib, and Stat3 inhibition permits the direct proapoptotic activity of sunitinib on tumor cells and positive effects on tumor immunologic microenvironment. Sunitinib 75-84 signal transducer and activator of transcription 3 Mus musculus 27-32 19276342-8 2009 More importantly, the expression of negative costimulatory molecules CTLA4 and PD-1 in both CD4 and CD8 T cells, and PDL-1 expression on MDSC and plasmacytoid dendritic cells, was also significantly decreased by sunitinib treatment. Sunitinib 212-221 cytotoxic T-lymphocyte protein 4 Sus scrofa 69-74 19276342-8 2009 More importantly, the expression of negative costimulatory molecules CTLA4 and PD-1 in both CD4 and CD8 T cells, and PDL-1 expression on MDSC and plasmacytoid dendritic cells, was also significantly decreased by sunitinib treatment. Sunitinib 212-221 CD4 molecule Sus scrofa 92-95 19244102-9 2009 These results suggest that Stat3 activity is important for RCC response to sunitinib, and Stat3 inhibition permits the direct proapoptotic activity of sunitinib on tumor cells and positive effects on tumor immunologic microenvironment. Sunitinib 151-160 signal transducer and activator of transcription 3 Mus musculus 90-95 19276342-8 2009 More importantly, the expression of negative costimulatory molecules CTLA4 and PD-1 in both CD4 and CD8 T cells, and PDL-1 expression on MDSC and plasmacytoid dendritic cells, was also significantly decreased by sunitinib treatment. Sunitinib 212-221 CD274 molecule Sus scrofa 117-122 19017755-6 2009 Patient was treated with sunitinib, a potent tyrosine kinase inhibitor of vascular endothelial growth factor, platelet-derived growth factor, RET, c-KIT, and FLT-3 receptors. Sunitinib 25-34 vascular endothelial growth factor A Homo sapiens 74-108 19035443-10 2009 Finally, we found significant growth suppressive effects of sunitinib in two acral melanoma cell lines; one harboring the D820Y mutation and one showing SCF-dependent KIT activation. Sunitinib 60-69 KIT ligand Homo sapiens 153-156 19035443-10 2009 Finally, we found significant growth suppressive effects of sunitinib in two acral melanoma cell lines; one harboring the D820Y mutation and one showing SCF-dependent KIT activation. Sunitinib 60-69 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 167-170 19035443-11 2009 These results show pathological activation of KIT in a substantial number of metastatic tumors of acral and mucosal melanomas, and suggest a potential therapeutic benefit of sunitinib for these melanomas. Sunitinib 174-183 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 46-49 19164557-0 2009 KIT kinase mutants show unique mechanisms of drug resistance to imatinib and sunitinib in gastrointestinal stromal tumor patients. Sunitinib 77-86 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 0-3 19164557-2 2009 The efficacy of the inhibitors imatinib mesylate and sunitinib malate in GIST patients has been linked to their inhibition of these mutant KIT proteins. Sunitinib 53-69 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 139-142 19164557-6 2009 Our results show that sunitinib targets the autoinhibited conformation of WT KIT and that the D816H mutant undergoes a shift in conformational equilibrium toward the active state. Sunitinib 22-31 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 77-80 18971320-0 2009 Sunitinib (Sutent, SU11248), a small-molecule receptor tyrosine kinase inhibitor, blocks function of the ATP-binding cassette (ABC) transporters P-glycoprotein (ABCB1) and ABCG2. Sunitinib 0-9 ATP binding cassette subfamily B member 1 Homo sapiens 145-159 18971320-0 2009 Sunitinib (Sutent, SU11248), a small-molecule receptor tyrosine kinase inhibitor, blocks function of the ATP-binding cassette (ABC) transporters P-glycoprotein (ABCB1) and ABCG2. Sunitinib 0-9 ATP binding cassette subfamily B member 1 Homo sapiens 161-166 18971320-0 2009 Sunitinib (Sutent, SU11248), a small-molecule receptor tyrosine kinase inhibitor, blocks function of the ATP-binding cassette (ABC) transporters P-glycoprotein (ABCB1) and ABCG2. Sunitinib 0-9 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 172-177 19284623-1 2009 BACKGROUND: Sunitinib is a protein tyrosine kinase-inhibitor targeting VEGFR, c-kit and PDGFR. Sunitinib 12-21 kinase insert domain receptor Homo sapiens 71-76 19284623-1 2009 BACKGROUND: Sunitinib is a protein tyrosine kinase-inhibitor targeting VEGFR, c-kit and PDGFR. Sunitinib 12-21 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 78-83 19284623-1 2009 BACKGROUND: Sunitinib is a protein tyrosine kinase-inhibitor targeting VEGFR, c-kit and PDGFR. Sunitinib 12-21 platelet derived growth factor receptor beta Homo sapiens 88-93 19127560-11 2009 CONCLUSIONS: The introduction of first-line sunitinib was associated with a doubling of overall survival compared with patients treated with IFN alone. Sunitinib 44-53 interferon alpha 1 Homo sapiens 141-144 18971320-0 2009 Sunitinib (Sutent, SU11248), a small-molecule receptor tyrosine kinase inhibitor, blocks function of the ATP-binding cassette (ABC) transporters P-glycoprotein (ABCB1) and ABCG2. Sunitinib 11-17 ATP binding cassette subfamily B member 1 Homo sapiens 145-159 18971320-0 2009 Sunitinib (Sutent, SU11248), a small-molecule receptor tyrosine kinase inhibitor, blocks function of the ATP-binding cassette (ABC) transporters P-glycoprotein (ABCB1) and ABCG2. Sunitinib 11-17 ATP binding cassette subfamily B member 1 Homo sapiens 161-166 18971320-0 2009 Sunitinib (Sutent, SU11248), a small-molecule receptor tyrosine kinase inhibitor, blocks function of the ATP-binding cassette (ABC) transporters P-glycoprotein (ABCB1) and ABCG2. Sunitinib 11-17 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 172-177 18971320-0 2009 Sunitinib (Sutent, SU11248), a small-molecule receptor tyrosine kinase inhibitor, blocks function of the ATP-binding cassette (ABC) transporters P-glycoprotein (ABCB1) and ABCG2. Sunitinib 19-26 ATP binding cassette subfamily B member 1 Homo sapiens 145-159 18971320-0 2009 Sunitinib (Sutent, SU11248), a small-molecule receptor tyrosine kinase inhibitor, blocks function of the ATP-binding cassette (ABC) transporters P-glycoprotein (ABCB1) and ABCG2. Sunitinib 19-26 ATP binding cassette subfamily B member 1 Homo sapiens 161-166 18971320-0 2009 Sunitinib (Sutent, SU11248), a small-molecule receptor tyrosine kinase inhibitor, blocks function of the ATP-binding cassette (ABC) transporters P-glycoprotein (ABCB1) and ABCG2. Sunitinib 19-26 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 172-177 18971320-3 2009 bioavailability and brain penetration of chemotherapy drugs in animal models, we sought to examine the effect of sunitinib on the ATP-binding cassette (ABC) drug transporters P-glycoprotein (P-gp, ABCB1), the multidrug resistance-associated protein 1 (ABCC1), and ABCG2, which are known to transport a wide variety of anticancer drugs. Sunitinib 113-122 ATP binding cassette subfamily B member 1 Homo sapiens 175-189 18971320-3 2009 bioavailability and brain penetration of chemotherapy drugs in animal models, we sought to examine the effect of sunitinib on the ATP-binding cassette (ABC) drug transporters P-glycoprotein (P-gp, ABCB1), the multidrug resistance-associated protein 1 (ABCC1), and ABCG2, which are known to transport a wide variety of anticancer drugs. Sunitinib 113-122 ATP binding cassette subfamily B member 1 Homo sapiens 191-195 18971320-3 2009 bioavailability and brain penetration of chemotherapy drugs in animal models, we sought to examine the effect of sunitinib on the ATP-binding cassette (ABC) drug transporters P-glycoprotein (P-gp, ABCB1), the multidrug resistance-associated protein 1 (ABCC1), and ABCG2, which are known to transport a wide variety of anticancer drugs. Sunitinib 113-122 ATP binding cassette subfamily B member 1 Homo sapiens 197-202 18971320-4 2009 In this study, we show that sunitinib inhibits P-gp- and ABCG2-mediated efflux of fluorescent substrates in cells overexpressing these transporters. Sunitinib 28-37 ATP binding cassette subfamily B member 1 Homo sapiens 47-51 18971320-4 2009 In this study, we show that sunitinib inhibits P-gp- and ABCG2-mediated efflux of fluorescent substrates in cells overexpressing these transporters. Sunitinib 28-37 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 57-62 18971320-5 2009 In 4-day cytotoxicity assays, at a nontoxic concentration (2 microM) sunitinib was able to partially reverse drug resistance mediated by P-gp and completely reverse resistance mediated by ABCG2. Sunitinib 69-78 ATP binding cassette subfamily B member 1 Homo sapiens 137-141 18971320-5 2009 In 4-day cytotoxicity assays, at a nontoxic concentration (2 microM) sunitinib was able to partially reverse drug resistance mediated by P-gp and completely reverse resistance mediated by ABCG2. Sunitinib 69-78 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 188-193 18971320-6 2009 We further show a direct interaction of sunitinib with the substrate binding pocket of these transporters as it inhibited binding of the photoaffinity substrate [(125)I]iodoarylazidoprazosin to P-gp (IC(50) = 14.2 microM) and ABCG2 (IC(50) = 1.33 microM). Sunitinib 40-49 ATP binding cassette subfamily B member 1 Homo sapiens 194-198 18971320-6 2009 We further show a direct interaction of sunitinib with the substrate binding pocket of these transporters as it inhibited binding of the photoaffinity substrate [(125)I]iodoarylazidoprazosin to P-gp (IC(50) = 14.2 microM) and ABCG2 (IC(50) = 1.33 microM). Sunitinib 40-49 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 226-231 18971320-8 2009 Conformation-sensitive antibody binding assays with the P-gp- and ABCG2-specific antibodies, UIC2 and 5D3, respectively, also confirmed the interaction of sunitinib with these transporters. Sunitinib 155-164 phosphoglycolate phosphatase Homo sapiens 56-61 18971320-8 2009 Conformation-sensitive antibody binding assays with the P-gp- and ABCG2-specific antibodies, UIC2 and 5D3, respectively, also confirmed the interaction of sunitinib with these transporters. Sunitinib 155-164 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 66-71 19017755-6 2009 Patient was treated with sunitinib, a potent tyrosine kinase inhibitor of vascular endothelial growth factor, platelet-derived growth factor, RET, c-KIT, and FLT-3 receptors. Sunitinib 25-34 ret proto-oncogene Homo sapiens 142-145 19017755-6 2009 Patient was treated with sunitinib, a potent tyrosine kinase inhibitor of vascular endothelial growth factor, platelet-derived growth factor, RET, c-KIT, and FLT-3 receptors. Sunitinib 25-34 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 147-152 19017755-6 2009 Patient was treated with sunitinib, a potent tyrosine kinase inhibitor of vascular endothelial growth factor, platelet-derived growth factor, RET, c-KIT, and FLT-3 receptors. Sunitinib 25-34 fms related receptor tyrosine kinase 3 Homo sapiens 158-163 19213665-1 2009 BACKGROUND: Sunitinib malate is an oral, multitargeted tyrosine kinase inhibitor that has demonstrated superior efficacy over interferon (IFN)-alpha in a phase III trial in first-line, metastatic renal cell carcinoma (RCC). Sunitinib 12-28 interferon alpha 1 Homo sapiens 126-148 19173737-11 2009 Using indirect comparisons with interferon-alpha as the common comparator, we found that sunitinib was superior to both sorafenib (HR 0.58, 95% CI, 0.38-0.86, P = < 0.001) and bevacizumab + IFN-a (HR 0.75, 95% CI, 0.60-0.93, P = 0.001). Sunitinib 89-98 interferon alpha 2 Homo sapiens 193-198 21686655-3 2009 Treatment with the novel multi-targeted tyrosine kinase inhibitor sunitinib resulted in a sustained partial response and reduced the level of the angiogenic marker PIGF. Sunitinib 66-75 phosphatidylinositol glycan anchor biosynthesis class F Homo sapiens 164-168 19054798-0 2009 Thrombotic microangiopathy secondary to VEGF pathway inhibition by sunitinib. Sunitinib 67-76 vascular endothelial growth factor A Homo sapiens 40-44 19054798-3 2009 It was shown recently that sunitinib, a small molecule inhibiting several tyrosine kinase receptors, including VEGF receptors, can also induce proteinuria, hypertension and biological features of TMA. Sunitinib 27-36 vascular endothelial growth factor A Homo sapiens 111-115 19212818-9 2009 In conclusion, opposite kinetics of two circulating CD34(bright) cell populations, HPCs and small CECs, were observed in sunitinib-treated RCC patients. Sunitinib 121-130 CD34 molecule Homo sapiens 52-56 19894779-0 2009 Sunitinib: a multitargeted receptor tyrosine kinase inhibitor in the era of molecular cancer therapies. Sunitinib 0-9 ret proto-oncogene Homo sapiens 27-51 20877672-10 2009 Sunitinib inhibits the vascular endothelial growth factor receptor (VEGFR), the platelet-derived growth factor receptor (PDGFR), c-kit and the colony-stimulating factor 1 (CSF-1) receptor. Sunitinib 0-9 kinase insert domain receptor Homo sapiens 23-66 20877672-10 2009 Sunitinib inhibits the vascular endothelial growth factor receptor (VEGFR), the platelet-derived growth factor receptor (PDGFR), c-kit and the colony-stimulating factor 1 (CSF-1) receptor. Sunitinib 0-9 kinase insert domain receptor Homo sapiens 68-73 20877672-10 2009 Sunitinib inhibits the vascular endothelial growth factor receptor (VEGFR), the platelet-derived growth factor receptor (PDGFR), c-kit and the colony-stimulating factor 1 (CSF-1) receptor. Sunitinib 0-9 platelet derived growth factor receptor beta Homo sapiens 80-119 20877672-10 2009 Sunitinib inhibits the vascular endothelial growth factor receptor (VEGFR), the platelet-derived growth factor receptor (PDGFR), c-kit and the colony-stimulating factor 1 (CSF-1) receptor. Sunitinib 0-9 platelet derived growth factor receptor beta Homo sapiens 121-126 20877672-10 2009 Sunitinib inhibits the vascular endothelial growth factor receptor (VEGFR), the platelet-derived growth factor receptor (PDGFR), c-kit and the colony-stimulating factor 1 (CSF-1) receptor. Sunitinib 0-9 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 129-134 20877672-10 2009 Sunitinib inhibits the vascular endothelial growth factor receptor (VEGFR), the platelet-derived growth factor receptor (PDGFR), c-kit and the colony-stimulating factor 1 (CSF-1) receptor. Sunitinib 0-9 colony stimulating factor 1 Homo sapiens 143-170 20877672-10 2009 Sunitinib inhibits the vascular endothelial growth factor receptor (VEGFR), the platelet-derived growth factor receptor (PDGFR), c-kit and the colony-stimulating factor 1 (CSF-1) receptor. Sunitinib 0-9 colony stimulating factor 1 receptor Homo sapiens 172-187 19096396-2 2009 In 2007, a pivotal phase III trial randomly allocated 750 patients with advanced renal cell carcinoma to receive either the VEGF-receptor tyrosine kinase inhibitor sunitinib or interferon alpha, and showed that sunitinib led to improved response rates, progression-free and overall survival. Sunitinib 211-220 vascular endothelial growth factor A Homo sapiens 124-128 19105714-1 2009 Sunitinib malate is an oral, multitargeted receptor tyrosine kinase inhibitor of VEGF receptors 1, 2 and 3; PDGF receptors alpha and beta, and other receptor tyrosine kinases implicated in tumor growth, angiogenesis and metastasis. Sunitinib 0-16 vascular endothelial growth factor A Homo sapiens 81-85 19096396-2 2009 In 2007, a pivotal phase III trial randomly allocated 750 patients with advanced renal cell carcinoma to receive either the VEGF-receptor tyrosine kinase inhibitor sunitinib or interferon alpha, and showed that sunitinib led to improved response rates, progression-free and overall survival. Sunitinib 164-173 vascular endothelial growth factor A Homo sapiens 124-128 19468197-6 2009 While such tyrosine kinase inhibitors, particularly those affecting RET activity such as vandetanib, sorafenib and sunitinib, are promising, the low rate of partial responses and absence of complete responses in all of the various trials of monotherapy emphasize the need for new and more effective single agents or combinations of therapeutic agents with acceptable toxicity. Sunitinib 115-124 ret proto-oncogene Homo sapiens 68-71 18618496-1 2008 BACKGROUND: Sunitinib and sorafenib are small molecules that inhibit the vascular endothelial growth factor and related receptors with substantial clinical activity reported in metastatic renal cell carcinoma (RCC). Sunitinib 12-21 vascular endothelial growth factor A Homo sapiens 73-107 19891126-5 2008 This review will summarize the major clinical trials and practical recommendations for the most studied VEGF inhibitors, including sunitinib, sorafenib, and bevacizumab; introduce novel VEGF inhibitor agents; outline side effects and toxicities; and discuss sequential and combination therapy with these agents. Sunitinib 131-140 vascular endothelial growth factor A Homo sapiens 104-108 18815214-3 2008 Sunitinib potently inhibited the enzyme activity of both AMP-activated protein kinase (AMPK) and the ribosomal S6 kinase RSK1 at therapeutically relevant concentrations. Sunitinib 0-9 protein kinase AMP-activated catalytic subunit alpha 2 Rattus norvegicus 57-85 18815214-3 2008 Sunitinib potently inhibited the enzyme activity of both AMP-activated protein kinase (AMPK) and the ribosomal S6 kinase RSK1 at therapeutically relevant concentrations. Sunitinib 0-9 protein kinase AMP-activated catalytic subunit alpha 2 Rattus norvegicus 87-91 18815214-3 2008 Sunitinib potently inhibited the enzyme activity of both AMP-activated protein kinase (AMPK) and the ribosomal S6 kinase RSK1 at therapeutically relevant concentrations. Sunitinib 0-9 ribosomal protein S6 kinase A1 Rattus norvegicus 121-125 18815214-6 2008 Sunitinib treatment also caused a dose-dependent reduction in myocyte protein levels of the phosphorylated alpha and beta isoforms of the AMPK phosphorylation target acetyl-Coenzyme A carboxylase. Sunitinib 0-9 protein kinase AMP-activated catalytic subunit alpha 2 Rattus norvegicus 138-142 18815214-11 2008 In conclusion, even though sunitinib potently inhibits AMPK and RSK1, given the extreme lack of kinase selectivity that sunitinib exhibits, it is likely that inhibition of other kinases or combinations of kinases are responsible for the cardiotoxic effects of sunitinib. Sunitinib 27-36 protein kinase AMP-activated catalytic subunit alpha 2 Rattus norvegicus 55-59 18815214-11 2008 In conclusion, even though sunitinib potently inhibits AMPK and RSK1, given the extreme lack of kinase selectivity that sunitinib exhibits, it is likely that inhibition of other kinases or combinations of kinases are responsible for the cardiotoxic effects of sunitinib. Sunitinib 27-36 ribosomal protein S6 kinase A1 Rattus norvegicus 64-68 18981453-0 2008 Nonclinical safety evaluation of sunitinib: a potent inhibitor of VEGF, PDGF, KIT, FLT3, and RET receptors. Sunitinib 33-42 vascular endothelial growth factor A Rattus norvegicus 66-70 18981453-0 2008 Nonclinical safety evaluation of sunitinib: a potent inhibitor of VEGF, PDGF, KIT, FLT3, and RET receptors. Sunitinib 33-42 KIT proto-oncogene receptor tyrosine kinase Rattus norvegicus 78-81 18981453-0 2008 Nonclinical safety evaluation of sunitinib: a potent inhibitor of VEGF, PDGF, KIT, FLT3, and RET receptors. Sunitinib 33-42 Fms related receptor tyrosine kinase 3 Rattus norvegicus 83-87 18932293-1 2008 The molecular targets of sunitinib are receptor tyrosine kinases (RTKs), and this drug has also been known to exert blocking effects on the activation of KIT, which is similar to the mechanism of action of imatinib. Sunitinib 25-34 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 154-157 18927310-4 2008 EXPERIMENTAL DESIGN: Type-1 (IFNgamma) and type-2 (interleukin-4) responses were assessed in T cells at baseline and day 28 of treatment with sunitinib (50 mg/d) by measuring intracellular cytokines after in vitro stimulation with anti-CD3/anti-CD28 antibodies. Sunitinib 142-151 interferon gamma Homo sapiens 29-37 18927310-4 2008 EXPERIMENTAL DESIGN: Type-1 (IFNgamma) and type-2 (interleukin-4) responses were assessed in T cells at baseline and day 28 of treatment with sunitinib (50 mg/d) by measuring intracellular cytokines after in vitro stimulation with anti-CD3/anti-CD28 antibodies. Sunitinib 142-151 interleukin 4 Homo sapiens 51-64 20020657-4 2009 We were the first to demonstrate that endothelial cell incubation was followed by increase in 5"-nucleotidase activity in the presence of sutent while celecoxib did not produce such effect. Sunitinib 138-144 5'-nucleotidase ecto Homo sapiens 94-109 19075590-4 2008 Both sunitinib and sorafenib target VEGF and PDGF receptor tyrosine kinases while bevacizumab is a monoclonal antibody to VEGF. Sunitinib 5-14 vascular endothelial growth factor A Homo sapiens 36-40 18955458-2 2008 Sunitinib, which targets KIT, PDGFRs, and several other kinases, has demonstrated efficacy in patients with GIST after they experience imatinib failure. Sunitinib 0-9 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 25-28 18955458-7 2008 RESULTS: Clinical benefit (partial response or stable disease for > or = 6 months) with sunitinib was observed for the three most common primary GIST genotypes: KIT exon 9 (58%), KIT exon 11 (34%), and wild-type KIT/PDGFRA (56%). Sunitinib 91-100 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 164-167 18955458-7 2008 RESULTS: Clinical benefit (partial response or stable disease for > or = 6 months) with sunitinib was observed for the three most common primary GIST genotypes: KIT exon 9 (58%), KIT exon 11 (34%), and wild-type KIT/PDGFRA (56%). Sunitinib 91-100 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 182-185 18955458-7 2008 RESULTS: Clinical benefit (partial response or stable disease for > or = 6 months) with sunitinib was observed for the three most common primary GIST genotypes: KIT exon 9 (58%), KIT exon 11 (34%), and wild-type KIT/PDGFRA (56%). Sunitinib 91-100 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 182-185 18955458-7 2008 RESULTS: Clinical benefit (partial response or stable disease for > or = 6 months) with sunitinib was observed for the three most common primary GIST genotypes: KIT exon 9 (58%), KIT exon 11 (34%), and wild-type KIT/PDGFRA (56%). Sunitinib 91-100 platelet derived growth factor receptor alpha Homo sapiens 219-225 18922106-2 2008 Sunitinib malate, sorafenib tosylate, bevacizumab +/- interferon-alfa, temsirolimus, and everolimus have improved clinical outcomes in randomized Phase III trials by inhibiting the VEGF and related pathways. Sunitinib 0-16 vascular endothelial growth factor A Homo sapiens 181-185 18374947-1 2008 OBJECTIVE: To determine the effects of sunitinib, a vascular endothelial growth factor receptor 2 (VEGFR-2) antagonist, on intra-abdominal adhesions. Sunitinib 39-48 kinase insert domain protein receptor Mus musculus 52-97 18794101-0 2008 Sunitinib-induced myeloid lineage redistribution in renal cell cancer patients: CD1c+ dendritic cell frequency predicts progression-free survival. Sunitinib 0-9 CD1c molecule Homo sapiens 80-84 18794101-9 2008 Subsequent to sunitinib treatment, an increase to high levels of myeloid DC (MDC) subset frequencies relative to other myeloid subsets, was specifically observed in patients experiencing tumor regression. Sunitinib 14-23 C-C motif chemokine ligand 22 Homo sapiens 65-75 18794101-9 2008 Subsequent to sunitinib treatment, an increase to high levels of myeloid DC (MDC) subset frequencies relative to other myeloid subsets, was specifically observed in patients experiencing tumor regression. Sunitinib 14-23 C-C motif chemokine ligand 22 Homo sapiens 77-80 18374947-1 2008 OBJECTIVE: To determine the effects of sunitinib, a vascular endothelial growth factor receptor 2 (VEGFR-2) antagonist, on intra-abdominal adhesions. Sunitinib 39-48 kinase insert domain protein receptor Mus musculus 99-106 18374947-4 2008 Sunitinib, a tyrosine kinase inhibitor with antiangiogenic and antitumor properties, may prevent or reduce postoperative abdominal adhesions by VEGFR-2 inhibition. Sunitinib 0-9 kinase insert domain protein receptor Mus musculus 144-151 18374947-14 2008 Sunitinib, a VEGFR-2 antagonist, significantly reduces adhesion formation in a murine model. Sunitinib 0-9 kinase insert domain protein receptor Mus musculus 13-20 18669461-8 2008 Mean plasma VEGF-A and PlGF levels significantly increased whereas VEGF-C and sVEGFR-3 levels decreased with sunitinib treatment. Sunitinib 109-118 vascular endothelial growth factor C Homo sapiens 67-73 18711190-7 2008 RESULTS: Treatment with sunitinib is associated with a gain in progression-free years of 0.41 and 0.35 over IFN-alpha and IL-2. Sunitinib 24-33 interferon alpha 1 Homo sapiens 108-117 18711190-7 2008 RESULTS: Treatment with sunitinib is associated with a gain in progression-free years of 0.41 and 0.35 over IFN-alpha and IL-2. Sunitinib 24-33 interleukin 2 Homo sapiens 122-126 18711190-9 2008 Both IFN-alpha and sunitinib treatments dominate IL-2 treatment; the incremental cost-effectiveness ratio of sunitinib versus IFN-alpha was $18,611 per progression-free year gained and $67,215 per LY gained, and the cost-utility ratio is $52,593 per QALY gained (at a 5% discount rate). Sunitinib 109-118 interferon alpha 1 Homo sapiens 5-14 18082353-10 2008 FINDINGS: Sunitinib (SU11248) is a novel multi-targeted tyrosine kinase inhibitor with activity not only against the receptor tyrosine kinase product of c-KIT but also other cell-signalling pathways that may be relevant in GIST; FLT3, platelet-derived growth receptor (PDGFR) and vascular endothelial growth factor receptor (VEGFR). Sunitinib 10-19 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 153-158 18082353-10 2008 FINDINGS: Sunitinib (SU11248) is a novel multi-targeted tyrosine kinase inhibitor with activity not only against the receptor tyrosine kinase product of c-KIT but also other cell-signalling pathways that may be relevant in GIST; FLT3, platelet-derived growth receptor (PDGFR) and vascular endothelial growth factor receptor (VEGFR). Sunitinib 10-19 fms related receptor tyrosine kinase 3 Homo sapiens 229-233 18082353-10 2008 FINDINGS: Sunitinib (SU11248) is a novel multi-targeted tyrosine kinase inhibitor with activity not only against the receptor tyrosine kinase product of c-KIT but also other cell-signalling pathways that may be relevant in GIST; FLT3, platelet-derived growth receptor (PDGFR) and vascular endothelial growth factor receptor (VEGFR). Sunitinib 10-19 platelet derived growth factor receptor beta Homo sapiens 269-274 18082353-10 2008 FINDINGS: Sunitinib (SU11248) is a novel multi-targeted tyrosine kinase inhibitor with activity not only against the receptor tyrosine kinase product of c-KIT but also other cell-signalling pathways that may be relevant in GIST; FLT3, platelet-derived growth receptor (PDGFR) and vascular endothelial growth factor receptor (VEGFR). Sunitinib 10-19 kinase insert domain receptor Homo sapiens 280-323 18082353-10 2008 FINDINGS: Sunitinib (SU11248) is a novel multi-targeted tyrosine kinase inhibitor with activity not only against the receptor tyrosine kinase product of c-KIT but also other cell-signalling pathways that may be relevant in GIST; FLT3, platelet-derived growth receptor (PDGFR) and vascular endothelial growth factor receptor (VEGFR). Sunitinib 10-19 kinase insert domain receptor Homo sapiens 325-330 18082353-10 2008 FINDINGS: Sunitinib (SU11248) is a novel multi-targeted tyrosine kinase inhibitor with activity not only against the receptor tyrosine kinase product of c-KIT but also other cell-signalling pathways that may be relevant in GIST; FLT3, platelet-derived growth receptor (PDGFR) and vascular endothelial growth factor receptor (VEGFR). Sunitinib 21-28 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 153-158 18082353-10 2008 FINDINGS: Sunitinib (SU11248) is a novel multi-targeted tyrosine kinase inhibitor with activity not only against the receptor tyrosine kinase product of c-KIT but also other cell-signalling pathways that may be relevant in GIST; FLT3, platelet-derived growth receptor (PDGFR) and vascular endothelial growth factor receptor (VEGFR). Sunitinib 21-28 fms related receptor tyrosine kinase 3 Homo sapiens 229-233 18082353-10 2008 FINDINGS: Sunitinib (SU11248) is a novel multi-targeted tyrosine kinase inhibitor with activity not only against the receptor tyrosine kinase product of c-KIT but also other cell-signalling pathways that may be relevant in GIST; FLT3, platelet-derived growth receptor (PDGFR) and vascular endothelial growth factor receptor (VEGFR). Sunitinib 21-28 platelet derived growth factor receptor beta Homo sapiens 269-274 18082353-10 2008 FINDINGS: Sunitinib (SU11248) is a novel multi-targeted tyrosine kinase inhibitor with activity not only against the receptor tyrosine kinase product of c-KIT but also other cell-signalling pathways that may be relevant in GIST; FLT3, platelet-derived growth receptor (PDGFR) and vascular endothelial growth factor receptor (VEGFR). Sunitinib 21-28 kinase insert domain receptor Homo sapiens 280-323 18082353-10 2008 FINDINGS: Sunitinib (SU11248) is a novel multi-targeted tyrosine kinase inhibitor with activity not only against the receptor tyrosine kinase product of c-KIT but also other cell-signalling pathways that may be relevant in GIST; FLT3, platelet-derived growth receptor (PDGFR) and vascular endothelial growth factor receptor (VEGFR). Sunitinib 21-28 kinase insert domain receptor Homo sapiens 325-330 18669461-11 2008 CONCLUSION: Sunitinib has substantial antitumor activity in patients with bevacizumab-refractory mRCC and modulates circulating VEGF pathway biomarkers. Sunitinib 12-21 vascular endothelial growth factor A Homo sapiens 128-132 18525301-13 2008 Further development of the thalidomide plus IL-2 combination therapy will address patients who have received molecular-targeted agents, such as sunitinib and sorafenib, as first- or second-line therapy. Sunitinib 144-153 interleukin 2 Homo sapiens 44-48 18648503-5 2008 Orally active anti-VEGF agents including sunitinib and ZM323881 effectively block hypoxia-induced retinal neovascularization. Sunitinib 41-50 vascular endothelial growth factor Aa Danio rerio 19-23 19296000-5 2008 Multi-kinase ihibitors (sorafenib and sunitinib) are orally administered small-molecules, that inhibit different receptors (essentials in the neoplastic angiogenesis), such as the VEGFR or PDGFR. Sunitinib 38-47 kinase insert domain receptor Homo sapiens 180-185 19296000-5 2008 Multi-kinase ihibitors (sorafenib and sunitinib) are orally administered small-molecules, that inhibit different receptors (essentials in the neoplastic angiogenesis), such as the VEGFR or PDGFR. Sunitinib 38-47 platelet derived growth factor receptor beta Homo sapiens 189-194 18514780-1 2008 PURPOSE: SU11248 (sunitinib) is a small-molecule tyrosine kinase inhibitor which targets VEGFR and PDGFR isoforms. Sunitinib 9-16 kinase insert domain receptor Homo sapiens 89-94 18514780-1 2008 PURPOSE: SU11248 (sunitinib) is a small-molecule tyrosine kinase inhibitor which targets VEGFR and PDGFR isoforms. Sunitinib 9-16 platelet derived growth factor receptor beta Homo sapiens 99-104 18514780-1 2008 PURPOSE: SU11248 (sunitinib) is a small-molecule tyrosine kinase inhibitor which targets VEGFR and PDGFR isoforms. Sunitinib 18-27 kinase insert domain receptor Homo sapiens 89-94 18514780-1 2008 PURPOSE: SU11248 (sunitinib) is a small-molecule tyrosine kinase inhibitor which targets VEGFR and PDGFR isoforms. Sunitinib 18-27 platelet derived growth factor receptor beta Homo sapiens 99-104 18514780-7 2008 RESULTS: SU11248 attenuated radiation-induced phosphorylation of Akt and ERK at 0, 5, 15, and 30 min. Sunitinib 9-16 AKT serine/threonine kinase 1 Homo sapiens 65-68 18514780-7 2008 RESULTS: SU11248 attenuated radiation-induced phosphorylation of Akt and ERK at 0, 5, 15, and 30 min. Sunitinib 9-16 EPH receptor B2 Homo sapiens 73-76 18293383-1 2008 BACKGROUND: Sunitinib is an orally bioavailable, multi-targeted tyrosine kinase inhibitor with selectivity for PDGF receptors, VEGF receptors, FLT3, and KIT. Sunitinib 12-21 FMS-like tyrosine kinase 3 Mus musculus 143-147 18293383-1 2008 BACKGROUND: Sunitinib is an orally bioavailable, multi-targeted tyrosine kinase inhibitor with selectivity for PDGF receptors, VEGF receptors, FLT3, and KIT. Sunitinib 12-21 KIT proto-oncogene receptor tyrosine kinase Mus musculus 153-156 18612155-3 2008 Sunitinib malate, an oral tyrosine kinase inhibitor, has activity against VEGFRs as well as platelet-derived growth factor receptors, stem-cell factor receptor, glial cell line-derived neurotrophic factor, and FMS-like tyrosine kinase-3. Sunitinib 0-16 fms related receptor tyrosine kinase 3 Homo sapiens 210-236 18418222-3 2008 Sunitinib is active against the RET kinase and has both antineoplastic and antiangiogenic properties. Sunitinib 0-9 ret proto-oncogene Homo sapiens 32-35 17983653-0 2008 Blockade of MEK/ERK signaling enhances sunitinib-induced growth inhibition and apoptosis of leukemia cells possessing activating mutations of the FLT3 gene. Sunitinib 39-48 mitogen-activated protein kinase kinase 7 Homo sapiens 12-15 17983653-0 2008 Blockade of MEK/ERK signaling enhances sunitinib-induced growth inhibition and apoptosis of leukemia cells possessing activating mutations of the FLT3 gene. Sunitinib 39-48 fms related receptor tyrosine kinase 3 Homo sapiens 146-150 17983653-7 2008 Interestingly, when AZD6244 was combined with sunitinib, a FLT3 kinase inhibitor, growth inhibition and apoptosis of both MV4-11 and MOLM13 cells were synergistically enhanced in association with further down-regulation of phospho-ERK1/2 and p-p70S6K in these cells. Sunitinib 46-55 fms related receptor tyrosine kinase 3 Homo sapiens 59-63 17983653-7 2008 Interestingly, when AZD6244 was combined with sunitinib, a FLT3 kinase inhibitor, growth inhibition and apoptosis of both MV4-11 and MOLM13 cells were synergistically enhanced in association with further down-regulation of phospho-ERK1/2 and p-p70S6K in these cells. Sunitinib 46-55 ribosomal protein S6 kinase B1 Homo sapiens 244-250 18537751-7 2008 Currently, the sole approved second-line drug is sunitinib--a multitargeted agent, an inhibitor of tyrosine kinase, of KIT and PDGFRA/B and of the vascular endothelial growth factor receptors (VEGFRs)-1, -2 and 3, FMS-like tyrosine kinase-3 (FLT3), colony stimulating factor 1 receptor (CSF-1R), and glial cell-line derived neurotrophic factor receptor (REarranged during Transfection; RET). Sunitinib 49-58 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 119-122 18537751-7 2008 Currently, the sole approved second-line drug is sunitinib--a multitargeted agent, an inhibitor of tyrosine kinase, of KIT and PDGFRA/B and of the vascular endothelial growth factor receptors (VEGFRs)-1, -2 and 3, FMS-like tyrosine kinase-3 (FLT3), colony stimulating factor 1 receptor (CSF-1R), and glial cell-line derived neurotrophic factor receptor (REarranged during Transfection; RET). Sunitinib 49-58 platelet derived growth factor receptor alpha Homo sapiens 127-133 18537751-7 2008 Currently, the sole approved second-line drug is sunitinib--a multitargeted agent, an inhibitor of tyrosine kinase, of KIT and PDGFRA/B and of the vascular endothelial growth factor receptors (VEGFRs)-1, -2 and 3, FMS-like tyrosine kinase-3 (FLT3), colony stimulating factor 1 receptor (CSF-1R), and glial cell-line derived neurotrophic factor receptor (REarranged during Transfection; RET). Sunitinib 49-58 fms related receptor tyrosine kinase 1 Homo sapiens 147-212 18537751-7 2008 Currently, the sole approved second-line drug is sunitinib--a multitargeted agent, an inhibitor of tyrosine kinase, of KIT and PDGFRA/B and of the vascular endothelial growth factor receptors (VEGFRs)-1, -2 and 3, FMS-like tyrosine kinase-3 (FLT3), colony stimulating factor 1 receptor (CSF-1R), and glial cell-line derived neurotrophic factor receptor (REarranged during Transfection; RET). Sunitinib 49-58 fms related receptor tyrosine kinase 3 Homo sapiens 214-240 18537751-7 2008 Currently, the sole approved second-line drug is sunitinib--a multitargeted agent, an inhibitor of tyrosine kinase, of KIT and PDGFRA/B and of the vascular endothelial growth factor receptors (VEGFRs)-1, -2 and 3, FMS-like tyrosine kinase-3 (FLT3), colony stimulating factor 1 receptor (CSF-1R), and glial cell-line derived neurotrophic factor receptor (REarranged during Transfection; RET). Sunitinib 49-58 fms related receptor tyrosine kinase 3 Homo sapiens 242-246 18537751-7 2008 Currently, the sole approved second-line drug is sunitinib--a multitargeted agent, an inhibitor of tyrosine kinase, of KIT and PDGFRA/B and of the vascular endothelial growth factor receptors (VEGFRs)-1, -2 and 3, FMS-like tyrosine kinase-3 (FLT3), colony stimulating factor 1 receptor (CSF-1R), and glial cell-line derived neurotrophic factor receptor (REarranged during Transfection; RET). Sunitinib 49-58 colony stimulating factor 1 receptor Homo sapiens 249-285 18537751-7 2008 Currently, the sole approved second-line drug is sunitinib--a multitargeted agent, an inhibitor of tyrosine kinase, of KIT and PDGFRA/B and of the vascular endothelial growth factor receptors (VEGFRs)-1, -2 and 3, FMS-like tyrosine kinase-3 (FLT3), colony stimulating factor 1 receptor (CSF-1R), and glial cell-line derived neurotrophic factor receptor (REarranged during Transfection; RET). Sunitinib 49-58 colony stimulating factor 1 receptor Homo sapiens 287-293 18537751-7 2008 Currently, the sole approved second-line drug is sunitinib--a multitargeted agent, an inhibitor of tyrosine kinase, of KIT and PDGFRA/B and of the vascular endothelial growth factor receptors (VEGFRs)-1, -2 and 3, FMS-like tyrosine kinase-3 (FLT3), colony stimulating factor 1 receptor (CSF-1R), and glial cell-line derived neurotrophic factor receptor (REarranged during Transfection; RET). Sunitinib 49-58 ret proto-oncogene Homo sapiens 386-389 18506667-9 2008 Small-molecule inhibitors of the VEGF receptor tyrosine kinase, such as sunitinib and sorafenib, have also shown promise in phase II trials and are being further investigated in phase III studies. Sunitinib 72-81 vascular endothelial growth factor A Homo sapiens 33-37 18483389-9 2008 In vitro studies showed that nilotinib, sunitinib, dasatinib, and sorafenib are more effective than imatinib against WT KIT. Sunitinib 40-49 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 120-123 18483300-3 2008 Sorafenib and sunitinib were potent inhibitors of cells with fms-like tyrosine kinase 3 internal tandem duplication (IC50, 2 and 7 nmol/L) and c-KIT N822K mutations (IC50, 23 and 40 nmol/L). Sunitinib 14-23 fms related receptor tyrosine kinase 3 Homo sapiens 61-87 18472378-2 2008 Among them, the major are those targeting the VEGF pathway, including anti-VEGF antibodies (bevacizumab) and VEGF receptor tyrosine kinase inhibitors (vatalanib, sorafenib, sunitinib...). Sunitinib 173-182 vascular endothelial growth factor A Homo sapiens 46-50 18483300-3 2008 Sorafenib and sunitinib were potent inhibitors of cells with fms-like tyrosine kinase 3 internal tandem duplication (IC50, 2 and 7 nmol/L) and c-KIT N822K mutations (IC50, 23 and 40 nmol/L). Sunitinib 14-23 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 143-148 18474019-4 2008 Sunitinib is a multitarget receptor tyrosine-kinase (TK) inhibitor, acting on VEGF receptor (VEGFR) and platelet-derived growth factor receptors (PDGFR). Sunitinib 0-9 kinase insert domain receptor Homo sapiens 78-91 18474019-4 2008 Sunitinib is a multitarget receptor tyrosine-kinase (TK) inhibitor, acting on VEGF receptor (VEGFR) and platelet-derived growth factor receptors (PDGFR). Sunitinib 0-9 kinase insert domain receptor Homo sapiens 93-98 18474019-4 2008 Sunitinib is a multitarget receptor tyrosine-kinase (TK) inhibitor, acting on VEGF receptor (VEGFR) and platelet-derived growth factor receptors (PDGFR). Sunitinib 0-9 platelet derived growth factor receptor beta Homo sapiens 104-144 18474019-4 2008 Sunitinib is a multitarget receptor tyrosine-kinase (TK) inhibitor, acting on VEGF receptor (VEGFR) and platelet-derived growth factor receptors (PDGFR). Sunitinib 0-9 platelet derived growth factor receptor beta Homo sapiens 146-151 17505827-1 2008 PURPOSE: Sunitinib, an oral multitargeted tyrosine kinase inhibitor that inhibits VEGFR, PDGFR, FLT3, KIT, and RET, is currently approved for the treatment of imatinib-refractory GIST and advanced renal cell carcinoma at a dose of 50 mg daily for 4 weeks followed by a 2-week off period (4/2 schedule). Sunitinib 9-18 kinase insert domain receptor Homo sapiens 82-87 18347007-1 2008 PURPOSE: Sunitinib is an oral, multitargeted tyrosine kinase inhibitor that inhibits vascular endothelial growth factor receptor (VEGFR), platelet-derived growth factor receptor, stem cell factor receptor (KIT), and colony-stimulating factor-1 receptor. Sunitinib 9-18 kinase insert domain receptor Homo sapiens 85-128 18347007-1 2008 PURPOSE: Sunitinib is an oral, multitargeted tyrosine kinase inhibitor that inhibits vascular endothelial growth factor receptor (VEGFR), platelet-derived growth factor receptor, stem cell factor receptor (KIT), and colony-stimulating factor-1 receptor. Sunitinib 9-18 kinase insert domain receptor Homo sapiens 130-135 18347007-1 2008 PURPOSE: Sunitinib is an oral, multitargeted tyrosine kinase inhibitor that inhibits vascular endothelial growth factor receptor (VEGFR), platelet-derived growth factor receptor, stem cell factor receptor (KIT), and colony-stimulating factor-1 receptor. Sunitinib 9-18 colony stimulating factor 1 receptor Homo sapiens 179-252 18265991-3 2008 Two tyrosine kinase inhibitors targeting the vascular endothelial growth factor (VEGF) receptor have been shown to improve the progression-free survival of patients in first-line (Sunitinib vs. interferon-alpha) or second-line treatment (Sorafenib vs. placebo). Sunitinib 180-189 vascular endothelial growth factor A Homo sapiens 81-85 18035517-3 2008 Two inhibitors targeting several protein kinases, including the VEGF receptor, have increased progression-free survival in patients with metastatic RCC and are now commercially available: sunitinib (Sutent) as first-line treatment and sorafenib (Nexavar) as second-line treatment. Sunitinib 188-197 vascular endothelial growth factor A Homo sapiens 64-68 18035517-3 2008 Two inhibitors targeting several protein kinases, including the VEGF receptor, have increased progression-free survival in patients with metastatic RCC and are now commercially available: sunitinib (Sutent) as first-line treatment and sorafenib (Nexavar) as second-line treatment. Sunitinib 199-205 vascular endothelial growth factor A Homo sapiens 64-68 17505827-1 2008 PURPOSE: Sunitinib, an oral multitargeted tyrosine kinase inhibitor that inhibits VEGFR, PDGFR, FLT3, KIT, and RET, is currently approved for the treatment of imatinib-refractory GIST and advanced renal cell carcinoma at a dose of 50 mg daily for 4 weeks followed by a 2-week off period (4/2 schedule). Sunitinib 9-18 platelet derived growth factor receptor beta Homo sapiens 89-94 17505827-1 2008 PURPOSE: Sunitinib, an oral multitargeted tyrosine kinase inhibitor that inhibits VEGFR, PDGFR, FLT3, KIT, and RET, is currently approved for the treatment of imatinib-refractory GIST and advanced renal cell carcinoma at a dose of 50 mg daily for 4 weeks followed by a 2-week off period (4/2 schedule). Sunitinib 9-18 fms related receptor tyrosine kinase 3 Homo sapiens 96-100 17505827-1 2008 PURPOSE: Sunitinib, an oral multitargeted tyrosine kinase inhibitor that inhibits VEGFR, PDGFR, FLT3, KIT, and RET, is currently approved for the treatment of imatinib-refractory GIST and advanced renal cell carcinoma at a dose of 50 mg daily for 4 weeks followed by a 2-week off period (4/2 schedule). Sunitinib 9-18 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 102-105 17505827-1 2008 PURPOSE: Sunitinib, an oral multitargeted tyrosine kinase inhibitor that inhibits VEGFR, PDGFR, FLT3, KIT, and RET, is currently approved for the treatment of imatinib-refractory GIST and advanced renal cell carcinoma at a dose of 50 mg daily for 4 weeks followed by a 2-week off period (4/2 schedule). Sunitinib 9-18 ret proto-oncogene Homo sapiens 111-114 18316579-6 2008 Furthermore, both sunitinib-treated groups maintained the molecular balance between angiopoietins Ang-1 and Ang-2, suggesting a critical role of angiopoietins in vascular normalization. Sunitinib 18-27 angiopoietin 1 Homo sapiens 98-103 18316579-6 2008 Furthermore, both sunitinib-treated groups maintained the molecular balance between angiopoietins Ang-1 and Ang-2, suggesting a critical role of angiopoietins in vascular normalization. Sunitinib 18-27 angiopoietin 2 Homo sapiens 108-113 17962201-2 2008 Sunitinib, also a VEGFR inhibitor, induces biochemical hypothyroidism in 85% of metastatic RCC patients, the majority of whom have signs or symptoms of hypothyroidism. Sunitinib 0-9 kinase insert domain receptor Homo sapiens 18-23 18287850-0 2008 F-18 FDG PET/CT assessment of gastrointestinal stromal tumor response to sunitinib malate therapy. Sunitinib 73-89 mastermind like domain containing 1 Homo sapiens 0-12 18366295-2 2008 In study populations comprising primarily good- and intermediate-risk patients with clear cell renal cell carcinoma and prior nephrectomy, prolonged progression-free survival was demonstrated for three angiogenesis-targeted agents: sunitinib (compared with interferon [IFN]), bevacizumab plus IFN (vs IFN alone) and sorafenib (vs placebo in cytokine-refractory patients). Sunitinib 232-241 interferon alpha 1 Homo sapiens 293-296 18366295-2 2008 In study populations comprising primarily good- and intermediate-risk patients with clear cell renal cell carcinoma and prior nephrectomy, prolonged progression-free survival was demonstrated for three angiogenesis-targeted agents: sunitinib (compared with interferon [IFN]), bevacizumab plus IFN (vs IFN alone) and sorafenib (vs placebo in cytokine-refractory patients). Sunitinib 232-241 interferon alpha 1 Homo sapiens 293-296 18230576-6 2008 Sunitinib is another approved drug and an inhibitor of multiple tyrosine kinases including KIT, PDGFR alpha as well as PDGFR beta and VEGFRs which are associated with angiogenesis. Sunitinib 0-9 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 91-94 17945482-1 2008 SU11248 sunitinib malate sutent is a selective inhibitor of certain protein tyrosine kinases including VEGF-R types 1-3 PDGF-R-a and -b, c-kit, and RET. Sunitinib 0-7 ret proto-oncogene Homo sapiens 148-151 18235126-2 2008 We evaluated the clinical activity and tolerability of sunitinib malate (SU11248), an oral, multitargeted tyrosine kinase inhibitor that blocks the activity of receptors for VEGF and PDGF, as well as related tyrosine kinases in patients with previously treated, advanced NSCLC. Sunitinib 55-71 vascular endothelial growth factor A Homo sapiens 174-178 18235126-2 2008 We evaluated the clinical activity and tolerability of sunitinib malate (SU11248), an oral, multitargeted tyrosine kinase inhibitor that blocks the activity of receptors for VEGF and PDGF, as well as related tyrosine kinases in patients with previously treated, advanced NSCLC. Sunitinib 73-80 vascular endothelial growth factor A Homo sapiens 174-178 18192256-2 2008 SUMMARY: Sunitinib malate is a potent inhibitor of vascular endothelial growth factor (VEGF) receptors, FMS-like tyrosine kinase 3 (FLT3), c-KIT, and platelet-derived growth factor (PDGF), which give the drug its direct antitumor and antiangiogenic properties. Sunitinib 9-25 vascular endothelial growth factor A Homo sapiens 51-85 18192256-2 2008 SUMMARY: Sunitinib malate is a potent inhibitor of vascular endothelial growth factor (VEGF) receptors, FMS-like tyrosine kinase 3 (FLT3), c-KIT, and platelet-derived growth factor (PDGF), which give the drug its direct antitumor and antiangiogenic properties. Sunitinib 9-25 vascular endothelial growth factor A Homo sapiens 87-91 18192256-2 2008 SUMMARY: Sunitinib malate is a potent inhibitor of vascular endothelial growth factor (VEGF) receptors, FMS-like tyrosine kinase 3 (FLT3), c-KIT, and platelet-derived growth factor (PDGF), which give the drug its direct antitumor and antiangiogenic properties. Sunitinib 9-25 fms related receptor tyrosine kinase 3 Homo sapiens 104-130 18192256-2 2008 SUMMARY: Sunitinib malate is a potent inhibitor of vascular endothelial growth factor (VEGF) receptors, FMS-like tyrosine kinase 3 (FLT3), c-KIT, and platelet-derived growth factor (PDGF), which give the drug its direct antitumor and antiangiogenic properties. Sunitinib 9-25 fms related receptor tyrosine kinase 3 Homo sapiens 132-136 18192256-2 2008 SUMMARY: Sunitinib malate is a potent inhibitor of vascular endothelial growth factor (VEGF) receptors, FMS-like tyrosine kinase 3 (FLT3), c-KIT, and platelet-derived growth factor (PDGF), which give the drug its direct antitumor and antiangiogenic properties. Sunitinib 9-25 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 139-144 18230576-6 2008 Sunitinib is another approved drug and an inhibitor of multiple tyrosine kinases including KIT, PDGFR alpha as well as PDGFR beta and VEGFRs which are associated with angiogenesis. Sunitinib 0-9 platelet derived growth factor receptor alpha Homo sapiens 96-107 18230576-6 2008 Sunitinib is another approved drug and an inhibitor of multiple tyrosine kinases including KIT, PDGFR alpha as well as PDGFR beta and VEGFRs which are associated with angiogenesis. Sunitinib 0-9 platelet derived growth factor receptor beta Homo sapiens 119-129 17942672-3 2007 However, here we report that an identical pattern of change is observed in normal nontumor-bearing mice treated with SU11248/sunitinib, a small-molecule inhibitor of VEGF and PDGF RTKs. Sunitinib 117-124 vascular endothelial growth factor A Mus musculus 166-170 18288997-10 2008 With the recent approval by FDA of Sorafenib and Sunitinib--targeting VEGFR, PDGFR, FLT-3 and c-Kit--a different scenario has been emerging, where a new generation of anti-cancer drugs, able to inhibit more than one pathway, would probably play a major role. Sunitinib 49-58 platelet derived growth factor receptor beta Homo sapiens 77-82 18288997-10 2008 With the recent approval by FDA of Sorafenib and Sunitinib--targeting VEGFR, PDGFR, FLT-3 and c-Kit--a different scenario has been emerging, where a new generation of anti-cancer drugs, able to inhibit more than one pathway, would probably play a major role. Sunitinib 49-58 fms related receptor tyrosine kinase 3 Homo sapiens 84-89 18288997-10 2008 With the recent approval by FDA of Sorafenib and Sunitinib--targeting VEGFR, PDGFR, FLT-3 and c-Kit--a different scenario has been emerging, where a new generation of anti-cancer drugs, able to inhibit more than one pathway, would probably play a major role. Sunitinib 49-58 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 94-99 17593391-10 2008 There were significantly higher pro-apoptotic effects (caspase-3 cleavage) observed for the sunitinib-docetaxel and sunitinib-docetaxel-cetuximab as compared to the other conditions. Sunitinib 92-101 caspase 3 Mus musculus 55-64 17593391-10 2008 There were significantly higher pro-apoptotic effects (caspase-3 cleavage) observed for the sunitinib-docetaxel and sunitinib-docetaxel-cetuximab as compared to the other conditions. Sunitinib 116-125 caspase 3 Mus musculus 55-64 18615363-3 2008 The new multitargeted kinase inhibitors sorafenib (Nexavar/BAY 43-9006) and sunitinib (Sutent/SUO 11248) interfere mainly with vascular endothelial growth factor and platelet-derived growth factor pathways. Sunitinib 76-85 vascular endothelial growth factor A Homo sapiens 127-161 18615363-3 2008 The new multitargeted kinase inhibitors sorafenib (Nexavar/BAY 43-9006) and sunitinib (Sutent/SUO 11248) interfere mainly with vascular endothelial growth factor and platelet-derived growth factor pathways. Sunitinib 87-93 vascular endothelial growth factor A Homo sapiens 127-161 17935226-5 2007 The in vivo effects of SU11248 treatment were monitored in the livers of cirrhotic rats by measuring angiogenesis, inflammatory infiltrate, fibrosis, alpha-smooth muscle actin (alpha-SMA) accumulation, differential gene expression by microarrays, and portal pressure. Sunitinib 23-30 actin gamma 2, smooth muscle Rattus norvegicus 150-175 17935226-5 2007 The in vivo effects of SU11248 treatment were monitored in the livers of cirrhotic rats by measuring angiogenesis, inflammatory infiltrate, fibrosis, alpha-smooth muscle actin (alpha-SMA) accumulation, differential gene expression by microarrays, and portal pressure. Sunitinib 23-30 actin gamma 2, smooth muscle Rattus norvegicus 177-186 17935226-9 2007 SU11248 treatment resulted in a significant decrease in hepatic vascular density, inflammatory infiltrate, alpha-SMA abundance, LX-2 viability, collagen expression, and portal pressure. Sunitinib 0-7 actin gamma 2, smooth muscle Rattus norvegicus 107-116 18165617-7 2007 Small-molecule multikinase inhibitors that target VEGF receptors (sunitinib and sorafenib) have a favorable toxicity profile and can prolong time to progression and preserve quality of life when used in newly diagnosed or previously treated patients. Sunitinib 66-75 vascular endothelial growth factor A Homo sapiens 50-54 18165647-10 2008 Two (4.8%) of 41 PRCC patients achieved a response (both patients were treated with sunitinib). Sunitinib 84-93 proline rich mitotic checkpoint control factor Homo sapiens 17-21 18165647-12 2008 Sunitinib-treated PRCC patients had a PFS of 11.9 months compared with 5.1 months for sorafenib-treated patients (P < .001). Sunitinib 0-9 proline rich mitotic checkpoint control factor Homo sapiens 18-22 18165647-13 2008 CONCLUSION: Patients with PRCC and ChRCC may have prolonged PFS from sunitinib and sorafenib, although clinical responses remain overall low in PRCC. Sunitinib 69-78 proline rich mitotic checkpoint control factor Homo sapiens 26-30 18272027-1 2007 Sunitinib is a tyrosine kinase inhibitor with activity against vascular endothelial growth factor (VEGF) receptor and platelet-derived growth factor receptor recently approved by the FDA for the treatment of advanced renal cell carcinoma (RCC). Sunitinib 0-9 vascular endothelial growth factor A Homo sapiens 99-103 17942672-3 2007 However, here we report that an identical pattern of change is observed in normal nontumor-bearing mice treated with SU11248/sunitinib, a small-molecule inhibitor of VEGF and PDGF RTKs. Sunitinib 125-134 vascular endothelial growth factor A Mus musculus 166-170 17622648-9 2007 Sunitinib treatment caused a 74% reduction in microvessel density (p < 0.05), an increase in tumor necrosis, and a decrease in number of GBM cells positive for MIB antibody. Sunitinib 0-9 MIB E3 ubiquitin protein ligase 1 Homo sapiens 163-166 17889720-0 2007 Sensitivity toward sorafenib and sunitinib varies between different activating and drug-resistant FLT3-ITD mutations. Sunitinib 33-42 fms related receptor tyrosine kinase 3 Homo sapiens 98-102 17889720-6 2007 Here we tested the efficacy of sunitinib and sorafenib to inhibit primary FLT3 activating mutations (ITD and D835Y) and of secondary resistance mutations. Sunitinib 31-40 fms related receptor tyrosine kinase 3 Homo sapiens 74-78 17889720-11 2007 Importantly, sensitivity toward sorafenib and sunitinib varies between the different secondary FLT3-ITD resistance mutations. Sunitinib 46-55 fms related receptor tyrosine kinase 3 Homo sapiens 95-99 17889720-12 2007 CONCLUSIONS: These results establish sensitivity profiles for the FLT3 inhibitors sunitinib and sorafenib. Sunitinib 82-91 fms related receptor tyrosine kinase 3 Homo sapiens 66-70 17761721-1 2007 Sunitinib malate is an oral, multitargeted tyrosine kinase inhibitor that targets both angiogenic pathways (i.e., vascular endothelial growth factor receptor and platelet-derived growth factor receptor) and direct pro-oncogenic pathways (e.g., stem-cell factor receptor and FMS-like tyrosine kinase-3). Sunitinib 0-16 fms related receptor tyrosine kinase 3 Homo sapiens 274-300 17701051-11 2007 Very recently, it has been shown that sunitinib may be especially effective in GISTs with KIT exon 9 mutation, whereas these tumors show only an intermediate response to imatinib. Sunitinib 38-47 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 90-93 17671153-2 2007 Most clinical trial data generated to date have been with either bevacizumab, a monoclonal antibody to VEGF, or small-molecule inhibitors of VEGF receptor (VEGFR) tyrosine kinase activity (sunitinib, sorafenib, and ZD6474). Sunitinib 189-198 kinase insert domain receptor Homo sapiens 156-161 17704140-1 2007 Two recent clinical trials have shown that the placenta growth factor (PlGF) is up-regulated after bevacizumab treatment in colorectal cancer and after SU11248 treatment in metastatic renal cell carcinoma. Sunitinib 152-159 placental growth factor Homo sapiens 47-69 17704140-1 2007 Two recent clinical trials have shown that the placenta growth factor (PlGF) is up-regulated after bevacizumab treatment in colorectal cancer and after SU11248 treatment in metastatic renal cell carcinoma. Sunitinib 152-159 placental growth factor Homo sapiens 71-75 17136543-2 2007 Sunitinib (sunitinib malate; SU11248; SUTENT; Pfizer Inc, New York, NY, USA) is a novel, orally bio-available, oxindole, multi-targeted tyrosine kinase inhibitor with high binding affinity for VEGFR and PDGFR which has shown anti-tumor and anti-angiogenic activities. Sunitinib 0-9 kinase insert domain receptor Homo sapiens 193-198 17136543-2 2007 Sunitinib (sunitinib malate; SU11248; SUTENT; Pfizer Inc, New York, NY, USA) is a novel, orally bio-available, oxindole, multi-targeted tyrosine kinase inhibitor with high binding affinity for VEGFR and PDGFR which has shown anti-tumor and anti-angiogenic activities. Sunitinib 0-9 platelet derived growth factor receptor beta Homo sapiens 203-208 17136543-2 2007 Sunitinib (sunitinib malate; SU11248; SUTENT; Pfizer Inc, New York, NY, USA) is a novel, orally bio-available, oxindole, multi-targeted tyrosine kinase inhibitor with high binding affinity for VEGFR and PDGFR which has shown anti-tumor and anti-angiogenic activities. Sunitinib 11-27 kinase insert domain receptor Homo sapiens 193-198 17136543-2 2007 Sunitinib (sunitinib malate; SU11248; SUTENT; Pfizer Inc, New York, NY, USA) is a novel, orally bio-available, oxindole, multi-targeted tyrosine kinase inhibitor with high binding affinity for VEGFR and PDGFR which has shown anti-tumor and anti-angiogenic activities. Sunitinib 11-27 platelet derived growth factor receptor beta Homo sapiens 203-208 17935273-7 2007 Sorafenib and sunitinib are synthetic, orally active agents shown to directly inhibit vascular endothelial growth factor receptors -2 and -3 (VEGFR-2, VEGFR-3) and platelet-derived growth factor receptor beta (PDGFR-beta), while temsirolimus is an mTOR inhibitor. Sunitinib 14-23 kinase insert domain receptor Homo sapiens 86-140 17935273-7 2007 Sorafenib and sunitinib are synthetic, orally active agents shown to directly inhibit vascular endothelial growth factor receptors -2 and -3 (VEGFR-2, VEGFR-3) and platelet-derived growth factor receptor beta (PDGFR-beta), while temsirolimus is an mTOR inhibitor. Sunitinib 14-23 kinase insert domain receptor Homo sapiens 142-149 17935273-7 2007 Sorafenib and sunitinib are synthetic, orally active agents shown to directly inhibit vascular endothelial growth factor receptors -2 and -3 (VEGFR-2, VEGFR-3) and platelet-derived growth factor receptor beta (PDGFR-beta), while temsirolimus is an mTOR inhibitor. Sunitinib 14-23 fms related receptor tyrosine kinase 4 Homo sapiens 151-158 17935273-7 2007 Sorafenib and sunitinib are synthetic, orally active agents shown to directly inhibit vascular endothelial growth factor receptors -2 and -3 (VEGFR-2, VEGFR-3) and platelet-derived growth factor receptor beta (PDGFR-beta), while temsirolimus is an mTOR inhibitor. Sunitinib 14-23 platelet derived growth factor receptor beta Homo sapiens 164-208 17935273-7 2007 Sorafenib and sunitinib are synthetic, orally active agents shown to directly inhibit vascular endothelial growth factor receptors -2 and -3 (VEGFR-2, VEGFR-3) and platelet-derived growth factor receptor beta (PDGFR-beta), while temsirolimus is an mTOR inhibitor. Sunitinib 14-23 platelet derived growth factor receptor beta Homo sapiens 210-220 17935273-7 2007 Sorafenib and sunitinib are synthetic, orally active agents shown to directly inhibit vascular endothelial growth factor receptors -2 and -3 (VEGFR-2, VEGFR-3) and platelet-derived growth factor receptor beta (PDGFR-beta), while temsirolimus is an mTOR inhibitor. Sunitinib 14-23 mechanistic target of rapamycin kinase Homo sapiens 248-252 17605814-0 2007 Circulating protein biomarkers of pharmacodynamic activity of sunitinib in patients with metastatic renal cell carcinoma: modulation of VEGF and VEGF-related proteins. Sunitinib 62-71 vascular endothelial growth factor A Homo sapiens 136-140 17710208-1 2007 Sunitinib (SU11248) is an orally bioavailable inhibitor that affects the receptor tyrosine kinases involved in tumour proliferation and angiogenesis, including vascular endothelial growth factor (VEGF) receptors 1, 2, 3, and platelet-derived growth factor receptors alpha (PDGFRA) and beta (PDGFRB). Sunitinib 0-9 fms related receptor tyrosine kinase 1 Homo sapiens 160-219 17710208-1 2007 Sunitinib (SU11248) is an orally bioavailable inhibitor that affects the receptor tyrosine kinases involved in tumour proliferation and angiogenesis, including vascular endothelial growth factor (VEGF) receptors 1, 2, 3, and platelet-derived growth factor receptors alpha (PDGFRA) and beta (PDGFRB). Sunitinib 0-9 platelet derived growth factor receptor alpha Homo sapiens 273-279 17710208-1 2007 Sunitinib (SU11248) is an orally bioavailable inhibitor that affects the receptor tyrosine kinases involved in tumour proliferation and angiogenesis, including vascular endothelial growth factor (VEGF) receptors 1, 2, 3, and platelet-derived growth factor receptors alpha (PDGFRA) and beta (PDGFRB). Sunitinib 0-9 platelet derived growth factor receptor beta Homo sapiens 291-297 17710208-1 2007 Sunitinib (SU11248) is an orally bioavailable inhibitor that affects the receptor tyrosine kinases involved in tumour proliferation and angiogenesis, including vascular endothelial growth factor (VEGF) receptors 1, 2, 3, and platelet-derived growth factor receptors alpha (PDGFRA) and beta (PDGFRB). Sunitinib 11-18 fms related receptor tyrosine kinase 1 Homo sapiens 160-219 17710208-1 2007 Sunitinib (SU11248) is an orally bioavailable inhibitor that affects the receptor tyrosine kinases involved in tumour proliferation and angiogenesis, including vascular endothelial growth factor (VEGF) receptors 1, 2, 3, and platelet-derived growth factor receptors alpha (PDGFRA) and beta (PDGFRB). Sunitinib 11-18 platelet derived growth factor receptor alpha Homo sapiens 273-279 17710208-1 2007 Sunitinib (SU11248) is an orally bioavailable inhibitor that affects the receptor tyrosine kinases involved in tumour proliferation and angiogenesis, including vascular endothelial growth factor (VEGF) receptors 1, 2, 3, and platelet-derived growth factor receptors alpha (PDGFRA) and beta (PDGFRB). Sunitinib 11-18 platelet derived growth factor receptor beta Homo sapiens 291-297 17710208-2 2007 Because angiogenesis is necessary for the growth and metastasis of solid tumours, and VEGF is believed to have a pivotal role in that process, SUNITINIB treatment may have broad-spectrum clinical utility. Sunitinib 143-152 vascular endothelial growth factor A Homo sapiens 86-90 17687201-4 2007 Meanwhile, sunitinib malate,which inhibits three VEGF-Rs and FLT 3 in addition to KIT as well as PDGF-R, was clinically evaluated in the phase II clinical trials for imatinib-resistant or intolerant GIST, and advanced renal cell carcinoma in Japan. Sunitinib 11-27 fms related receptor tyrosine kinase 3 Homo sapiens 61-66 17687201-4 2007 Meanwhile, sunitinib malate,which inhibits three VEGF-Rs and FLT 3 in addition to KIT as well as PDGF-R, was clinically evaluated in the phase II clinical trials for imatinib-resistant or intolerant GIST, and advanced renal cell carcinoma in Japan. Sunitinib 11-27 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 82-85 17657252-5 2007 Sunitinib, sorafenib and axitinib are kinase inhibitors that inhibit the VEGF, platelet-derived growth factor and c-kit receptor tyrosine kinases. Sunitinib 0-9 vascular endothelial growth factor A Homo sapiens 73-77 17655513-6 2007 Sorafenib and sunitinib are novel therapies that target growth factor receptors known to be activated by the hypoxia-inducible factor and the Ras-Raf/MEK/ERK pathways. Sunitinib 14-23 zinc fingers and homeoboxes 2 Homo sapiens 146-149 17655513-6 2007 Sorafenib and sunitinib are novel therapies that target growth factor receptors known to be activated by the hypoxia-inducible factor and the Ras-Raf/MEK/ERK pathways. Sunitinib 14-23 mitogen-activated protein kinase kinase 7 Homo sapiens 150-153 17655513-6 2007 Sorafenib and sunitinib are novel therapies that target growth factor receptors known to be activated by the hypoxia-inducible factor and the Ras-Raf/MEK/ERK pathways. Sunitinib 14-23 mitogen-activated protein kinase 1 Homo sapiens 154-157 17605814-0 2007 Circulating protein biomarkers of pharmacodynamic activity of sunitinib in patients with metastatic renal cell carcinoma: modulation of VEGF and VEGF-related proteins. Sunitinib 62-71 vascular endothelial growth factor A Homo sapiens 145-149 17605814-9 2007 CONCLUSION: Sunitinib treatment in advanced RCC patients leads to modulation of plasma levels of circulating proteins involved in VEGF signaling, including soluble forms of two VEGF receptors. Sunitinib 12-21 vascular endothelial growth factor A Homo sapiens 130-134 17605814-9 2007 CONCLUSION: Sunitinib treatment in advanced RCC patients leads to modulation of plasma levels of circulating proteins involved in VEGF signaling, including soluble forms of two VEGF receptors. Sunitinib 12-21 vascular endothelial growth factor A Homo sapiens 177-181 17588358-2 2007 Sunitinib is a tyrosine kinase inhibitor with a wide range of kinase inhibition, including KIT, platelet-derived growth factor receptor (PDGFR), vascular endothelial growth factor (VEGF), and FLT3. Sunitinib 0-9 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 91-94 17588358-2 2007 Sunitinib is a tyrosine kinase inhibitor with a wide range of kinase inhibition, including KIT, platelet-derived growth factor receptor (PDGFR), vascular endothelial growth factor (VEGF), and FLT3. Sunitinib 0-9 platelet derived growth factor receptor beta Homo sapiens 96-135 17825686-8 2007 Sunitinib is metabolized by cytochrome P450 (CYP) 3A4 to an active metabolite, SU12662, which is further metabolized by CYP3A4 to an inactive moiety. Sunitinib 0-9 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 120-126 17588358-2 2007 Sunitinib is a tyrosine kinase inhibitor with a wide range of kinase inhibition, including KIT, platelet-derived growth factor receptor (PDGFR), vascular endothelial growth factor (VEGF), and FLT3. Sunitinib 0-9 platelet derived growth factor receptor beta Homo sapiens 137-142 17588358-2 2007 Sunitinib is a tyrosine kinase inhibitor with a wide range of kinase inhibition, including KIT, platelet-derived growth factor receptor (PDGFR), vascular endothelial growth factor (VEGF), and FLT3. Sunitinib 0-9 vascular endothelial growth factor A Homo sapiens 145-179 17588358-2 2007 Sunitinib is a tyrosine kinase inhibitor with a wide range of kinase inhibition, including KIT, platelet-derived growth factor receptor (PDGFR), vascular endothelial growth factor (VEGF), and FLT3. Sunitinib 0-9 vascular endothelial growth factor A Homo sapiens 181-185 17588358-2 2007 Sunitinib is a tyrosine kinase inhibitor with a wide range of kinase inhibition, including KIT, platelet-derived growth factor receptor (PDGFR), vascular endothelial growth factor (VEGF), and FLT3. Sunitinib 0-9 fms related receptor tyrosine kinase 3 Homo sapiens 192-196 17552016-0 2007 Effect of sunitinib on metastatic gastrointestinal stromal tumor in patients with neurofibromatosis type 1: a case report. Sunitinib 10-19 neurofibromin 1 Homo sapiens 82-106 17545799-8 2007 Patients with gastrointestinal stromal tumor with KIT exon 9 mutation may benefit more from sunitinib than those with exon 11 mutation. Sunitinib 92-101 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 50-53 17591808-8 2007 More recently Sunitinib, a new KIT/PDGFRA kinase inhibitor, has been tested in patients with GIST resistant to imatinib, with promising results. Sunitinib 14-23 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 31-34 17591808-8 2007 More recently Sunitinib, a new KIT/PDGFRA kinase inhibitor, has been tested in patients with GIST resistant to imatinib, with promising results. Sunitinib 14-23 platelet derived growth factor receptor alpha Homo sapiens 35-41 17591828-6 2007 Sunitinib targets selectively vascular endothelial growth factor, KIT, Flt3 and platelet-derived growth factor receptors and the receptor encoded by the ret proto-oncogene. Sunitinib 0-9 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 66-69 17591828-6 2007 Sunitinib targets selectively vascular endothelial growth factor, KIT, Flt3 and platelet-derived growth factor receptors and the receptor encoded by the ret proto-oncogene. Sunitinib 0-9 fms related receptor tyrosine kinase 3 Homo sapiens 71-75 17552016-14 2007 The metastatic lesions in the liver and omentum were decreased in size after four courses, suggesting that sunitinib is also an effective treatment modality for metastatic GIST in NF patients. Sunitinib 107-116 neurofascin Homo sapiens 180-182 17355219-3 2007 One of the most clear examples is the identification of KIT and platelet-derived growth factor receptor-alpha kinase mutations in gastrointestinal stromal tumours, a subset of sarcomas arising from precursors of the interstitial cells of Cajal in the digestive tract, which led to the development of imatinib, sunitinib and other tyrosine kinase inhibitors for the treatment of solid tumours. Sunitinib 310-319 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 56-59 17392388-3 2007 Several anti-VEGF agents, including ligand-binding agents such as bevacizumab and the small molecule inhibitors of VEGF and related receptors such as sunitinib and sorafenib, have demonstrated clinical activity in patients with metastatic RCC. Sunitinib 150-159 vascular endothelial growth factor A Homo sapiens 13-17 17392388-3 2007 Several anti-VEGF agents, including ligand-binding agents such as bevacizumab and the small molecule inhibitors of VEGF and related receptors such as sunitinib and sorafenib, have demonstrated clinical activity in patients with metastatic RCC. Sunitinib 150-159 vascular endothelial growth factor A Homo sapiens 115-119 17272980-6 2007 Sunitinib is a vascular endothelial growth factor receptor 1, 2, and 3, c-KIT, and platelet-derived growth factor receptor alpha and beta tyrosine kinase inhibitor. Sunitinib 0-9 fms related receptor tyrosine kinase 1 Homo sapiens 15-70 17272980-6 2007 Sunitinib is a vascular endothelial growth factor receptor 1, 2, and 3, c-KIT, and platelet-derived growth factor receptor alpha and beta tyrosine kinase inhibitor. Sunitinib 0-9 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 72-77 17272980-6 2007 Sunitinib is a vascular endothelial growth factor receptor 1, 2, and 3, c-KIT, and platelet-derived growth factor receptor alpha and beta tyrosine kinase inhibitor. Sunitinib 0-9 platelet derived growth factor receptor alpha Homo sapiens 83-128 17367763-0 2007 Sunitinib: a VEGF and PDGF receptor protein kinase and angiogenesis inhibitor. Sunitinib 0-9 vascular endothelial growth factor A Homo sapiens 13-17 17367763-1 2007 Sunitinib (SU-11248, Sutent) inhibits at least eight receptor protein-tyrosine kinases including vascular endothelial growth factor receptors 1-3 (VEGFR1-VEGFR3), platelet-derived growth factor receptors (PDGFRalpha and PDGFRbeta), stem cell factor receptor (Kit), Flt-3, and colony-stimulating factor-1 receptor (CSF-1R). Sunitinib 0-9 fms related receptor tyrosine kinase 1 Homo sapiens 97-145 17367763-1 2007 Sunitinib (SU-11248, Sutent) inhibits at least eight receptor protein-tyrosine kinases including vascular endothelial growth factor receptors 1-3 (VEGFR1-VEGFR3), platelet-derived growth factor receptors (PDGFRalpha and PDGFRbeta), stem cell factor receptor (Kit), Flt-3, and colony-stimulating factor-1 receptor (CSF-1R). Sunitinib 0-9 fms related receptor tyrosine kinase 1 Homo sapiens 147-153 17367763-1 2007 Sunitinib (SU-11248, Sutent) inhibits at least eight receptor protein-tyrosine kinases including vascular endothelial growth factor receptors 1-3 (VEGFR1-VEGFR3), platelet-derived growth factor receptors (PDGFRalpha and PDGFRbeta), stem cell factor receptor (Kit), Flt-3, and colony-stimulating factor-1 receptor (CSF-1R). Sunitinib 0-9 fms related receptor tyrosine kinase 4 Homo sapiens 154-160 17367763-1 2007 Sunitinib (SU-11248, Sutent) inhibits at least eight receptor protein-tyrosine kinases including vascular endothelial growth factor receptors 1-3 (VEGFR1-VEGFR3), platelet-derived growth factor receptors (PDGFRalpha and PDGFRbeta), stem cell factor receptor (Kit), Flt-3, and colony-stimulating factor-1 receptor (CSF-1R). Sunitinib 0-9 platelet derived growth factor receptor alpha Homo sapiens 205-215 17367763-1 2007 Sunitinib (SU-11248, Sutent) inhibits at least eight receptor protein-tyrosine kinases including vascular endothelial growth factor receptors 1-3 (VEGFR1-VEGFR3), platelet-derived growth factor receptors (PDGFRalpha and PDGFRbeta), stem cell factor receptor (Kit), Flt-3, and colony-stimulating factor-1 receptor (CSF-1R). Sunitinib 0-9 platelet derived growth factor receptor beta Homo sapiens 220-229 17367763-1 2007 Sunitinib (SU-11248, Sutent) inhibits at least eight receptor protein-tyrosine kinases including vascular endothelial growth factor receptors 1-3 (VEGFR1-VEGFR3), platelet-derived growth factor receptors (PDGFRalpha and PDGFRbeta), stem cell factor receptor (Kit), Flt-3, and colony-stimulating factor-1 receptor (CSF-1R). Sunitinib 0-9 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 259-262 17367763-1 2007 Sunitinib (SU-11248, Sutent) inhibits at least eight receptor protein-tyrosine kinases including vascular endothelial growth factor receptors 1-3 (VEGFR1-VEGFR3), platelet-derived growth factor receptors (PDGFRalpha and PDGFRbeta), stem cell factor receptor (Kit), Flt-3, and colony-stimulating factor-1 receptor (CSF-1R). Sunitinib 0-9 fms related receptor tyrosine kinase 3 Homo sapiens 265-270 17367763-1 2007 Sunitinib (SU-11248, Sutent) inhibits at least eight receptor protein-tyrosine kinases including vascular endothelial growth factor receptors 1-3 (VEGFR1-VEGFR3), platelet-derived growth factor receptors (PDGFRalpha and PDGFRbeta), stem cell factor receptor (Kit), Flt-3, and colony-stimulating factor-1 receptor (CSF-1R). Sunitinib 0-9 colony stimulating factor 1 receptor Homo sapiens 276-312 17367763-1 2007 Sunitinib (SU-11248, Sutent) inhibits at least eight receptor protein-tyrosine kinases including vascular endothelial growth factor receptors 1-3 (VEGFR1-VEGFR3), platelet-derived growth factor receptors (PDGFRalpha and PDGFRbeta), stem cell factor receptor (Kit), Flt-3, and colony-stimulating factor-1 receptor (CSF-1R). Sunitinib 0-9 colony stimulating factor 1 receptor Homo sapiens 314-321 17367763-1 2007 Sunitinib (SU-11248, Sutent) inhibits at least eight receptor protein-tyrosine kinases including vascular endothelial growth factor receptors 1-3 (VEGFR1-VEGFR3), platelet-derived growth factor receptors (PDGFRalpha and PDGFRbeta), stem cell factor receptor (Kit), Flt-3, and colony-stimulating factor-1 receptor (CSF-1R). Sunitinib 11-19 fms related receptor tyrosine kinase 1 Homo sapiens 97-145 17367763-1 2007 Sunitinib (SU-11248, Sutent) inhibits at least eight receptor protein-tyrosine kinases including vascular endothelial growth factor receptors 1-3 (VEGFR1-VEGFR3), platelet-derived growth factor receptors (PDGFRalpha and PDGFRbeta), stem cell factor receptor (Kit), Flt-3, and colony-stimulating factor-1 receptor (CSF-1R). Sunitinib 11-19 fms related receptor tyrosine kinase 1 Homo sapiens 147-153 17367763-1 2007 Sunitinib (SU-11248, Sutent) inhibits at least eight receptor protein-tyrosine kinases including vascular endothelial growth factor receptors 1-3 (VEGFR1-VEGFR3), platelet-derived growth factor receptors (PDGFRalpha and PDGFRbeta), stem cell factor receptor (Kit), Flt-3, and colony-stimulating factor-1 receptor (CSF-1R). Sunitinib 21-27 fms related receptor tyrosine kinase 1 Homo sapiens 97-145 17367763-1 2007 Sunitinib (SU-11248, Sutent) inhibits at least eight receptor protein-tyrosine kinases including vascular endothelial growth factor receptors 1-3 (VEGFR1-VEGFR3), platelet-derived growth factor receptors (PDGFRalpha and PDGFRbeta), stem cell factor receptor (Kit), Flt-3, and colony-stimulating factor-1 receptor (CSF-1R). Sunitinib 21-27 fms related receptor tyrosine kinase 1 Homo sapiens 147-153 17367763-1 2007 Sunitinib (SU-11248, Sutent) inhibits at least eight receptor protein-tyrosine kinases including vascular endothelial growth factor receptors 1-3 (VEGFR1-VEGFR3), platelet-derived growth factor receptors (PDGFRalpha and PDGFRbeta), stem cell factor receptor (Kit), Flt-3, and colony-stimulating factor-1 receptor (CSF-1R). Sunitinib 21-27 fms related receptor tyrosine kinase 4 Homo sapiens 154-160 17367763-4 2007 Sunitinib inhibits angiogenesis by diminishing signaling through VEGFR1, VEGFR2, and PDGFRbeta. Sunitinib 0-9 fms related receptor tyrosine kinase 1 Homo sapiens 65-71 17367763-4 2007 Sunitinib inhibits angiogenesis by diminishing signaling through VEGFR1, VEGFR2, and PDGFRbeta. Sunitinib 0-9 kinase insert domain receptor Homo sapiens 73-79 17367763-4 2007 Sunitinib inhibits angiogenesis by diminishing signaling through VEGFR1, VEGFR2, and PDGFRbeta. Sunitinib 0-9 platelet derived growth factor receptor beta Homo sapiens 85-94 17367763-7 2007 Sunitinib is approved for the treatment of those tumors that are resistant to imatinib (STI-571, Gleevec), another Kit and PDGFRalpha protein-tyrosine kinase inhibitor. Sunitinib 0-9 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 115-118 17367763-7 2007 Sunitinib is approved for the treatment of those tumors that are resistant to imatinib (STI-571, Gleevec), another Kit and PDGFRalpha protein-tyrosine kinase inhibitor. Sunitinib 0-9 platelet derived growth factor receptor alpha Homo sapiens 123-133 17573273-2 2007 This article will review the pathology of GISTs, the molecular pathology related to c-kit, and disease management and will discuss a new drug approved in the management of GISTs, sunitinib malate. Sunitinib 179-195 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 84-89 17619763-3 2007 Sutent and sorafenib are orally available inhibitors of the VEGFR and platelet derived growth factor receptor (PDGFR). Sunitinib 0-6 kinase insert domain receptor Homo sapiens 60-65 17619763-3 2007 Sutent and sorafenib are orally available inhibitors of the VEGFR and platelet derived growth factor receptor (PDGFR). Sunitinib 0-6 platelet derived growth factor receptor beta Homo sapiens 70-109 17619763-3 2007 Sutent and sorafenib are orally available inhibitors of the VEGFR and platelet derived growth factor receptor (PDGFR). Sunitinib 0-6 platelet derived growth factor receptor beta Homo sapiens 111-116 17355219-3 2007 One of the most clear examples is the identification of KIT and platelet-derived growth factor receptor-alpha kinase mutations in gastrointestinal stromal tumours, a subset of sarcomas arising from precursors of the interstitial cells of Cajal in the digestive tract, which led to the development of imatinib, sunitinib and other tyrosine kinase inhibitors for the treatment of solid tumours. Sunitinib 310-319 platelet derived growth factor receptor alpha Homo sapiens 64-109 17327610-2 2007 Tyrosine kinase inhibitors such as SU6668 and SU5416 (semaxanib) demonstrated poor pharmacologic properties and limited efficacy; therefore, sunitinib was rationally designed and chosen for its high bioavailability and its nanomolar-range potency against the antiangiogenic receptor tyrosine kinases (RTKs)--vascular endothelial growth factor receptor (VEGFR) and platelet-derived growth factor receptor (PDGFR). Sunitinib 141-150 kinase insert domain receptor Homo sapiens 353-358 17327610-2 2007 Tyrosine kinase inhibitors such as SU6668 and SU5416 (semaxanib) demonstrated poor pharmacologic properties and limited efficacy; therefore, sunitinib was rationally designed and chosen for its high bioavailability and its nanomolar-range potency against the antiangiogenic receptor tyrosine kinases (RTKs)--vascular endothelial growth factor receptor (VEGFR) and platelet-derived growth factor receptor (PDGFR). Sunitinib 141-150 platelet derived growth factor receptor beta Homo sapiens 364-403 17327610-2 2007 Tyrosine kinase inhibitors such as SU6668 and SU5416 (semaxanib) demonstrated poor pharmacologic properties and limited efficacy; therefore, sunitinib was rationally designed and chosen for its high bioavailability and its nanomolar-range potency against the antiangiogenic receptor tyrosine kinases (RTKs)--vascular endothelial growth factor receptor (VEGFR) and platelet-derived growth factor receptor (PDGFR). Sunitinib 141-150 platelet derived growth factor receptor beta Homo sapiens 405-410 17327610-3 2007 Sunitinib inhibits other tyrosine kinases including, KIT, FLT3, colony-stimulating factor 1 (CSF-1), and RET, which are involved in a number of malignancies including small-cell lung cancer, GI stromal tumors (GISTs), breast cancer, acute myelogenous leukemia, multiple endocrine neoplasia types 2A and 2B, and familial medullary thyroid carcinoma. Sunitinib 0-9 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 53-56 17327610-3 2007 Sunitinib inhibits other tyrosine kinases including, KIT, FLT3, colony-stimulating factor 1 (CSF-1), and RET, which are involved in a number of malignancies including small-cell lung cancer, GI stromal tumors (GISTs), breast cancer, acute myelogenous leukemia, multiple endocrine neoplasia types 2A and 2B, and familial medullary thyroid carcinoma. Sunitinib 0-9 fms related receptor tyrosine kinase 3 Homo sapiens 58-62 17327610-3 2007 Sunitinib inhibits other tyrosine kinases including, KIT, FLT3, colony-stimulating factor 1 (CSF-1), and RET, which are involved in a number of malignancies including small-cell lung cancer, GI stromal tumors (GISTs), breast cancer, acute myelogenous leukemia, multiple endocrine neoplasia types 2A and 2B, and familial medullary thyroid carcinoma. Sunitinib 0-9 colony stimulating factor 1 Homo sapiens 64-91 17327610-3 2007 Sunitinib inhibits other tyrosine kinases including, KIT, FLT3, colony-stimulating factor 1 (CSF-1), and RET, which are involved in a number of malignancies including small-cell lung cancer, GI stromal tumors (GISTs), breast cancer, acute myelogenous leukemia, multiple endocrine neoplasia types 2A and 2B, and familial medullary thyroid carcinoma. Sunitinib 0-9 colony stimulating factor 1 Homo sapiens 93-98 17327610-3 2007 Sunitinib inhibits other tyrosine kinases including, KIT, FLT3, colony-stimulating factor 1 (CSF-1), and RET, which are involved in a number of malignancies including small-cell lung cancer, GI stromal tumors (GISTs), breast cancer, acute myelogenous leukemia, multiple endocrine neoplasia types 2A and 2B, and familial medullary thyroid carcinoma. Sunitinib 0-9 ret proto-oncogene Homo sapiens 105-108 17327610-4 2007 Sunitinib demonstrated robust antitumor activity in preclinical studies resulting not only in tumor growth inhibition, but tumor regression in models of colon cancer, non-small-cell lung cancer, melanoma, renal carcinoma, and squamous cell carcinoma, which were associated with inhibition of VEGFR and PDGFR phosphorylation. Sunitinib 0-9 kinase insert domain receptor Homo sapiens 292-297 17327610-4 2007 Sunitinib demonstrated robust antitumor activity in preclinical studies resulting not only in tumor growth inhibition, but tumor regression in models of colon cancer, non-small-cell lung cancer, melanoma, renal carcinoma, and squamous cell carcinoma, which were associated with inhibition of VEGFR and PDGFR phosphorylation. Sunitinib 0-9 platelet derived growth factor receptor beta Homo sapiens 302-307 17208434-2 2007 These rare neoplasms are remarkably sensitive to the KIT and PDGFRA kinase inhibitors imatinib (also known as Gleevec) and sunitinib (Sutent), which have recently been approved as the standard therapeutic courses for patients with inoperable GIST. Sunitinib 134-140 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 53-56 17621562-6 2007 A recent Phase III study evaluating sunitinib as first-line therapy showed a significant difference when compared to interferonalfa (IFN-alpha) for a progression-free survival of 11 months in the sunitinib arm and 5 months in the IFN-alpha arm (hazard ratio 0.42; 95% CI 0.32-0.54; P50.001). Sunitinib 36-45 interferon alpha 1 Homo sapiens 230-239 17317817-3 2007 Three agents targeting the VEGF pathway have shown clinical activity as monotherapy in metastatic renal cell carcinoma: the anti-VEGF monoclonal antibody, bevacizumab, and small-molecule VEGF receptor tyrosine kinase inhibitors, sorafenib and sunitinib. Sunitinib 243-252 vascular endothelial growth factor A Homo sapiens 27-31 17208434-2 2007 These rare neoplasms are remarkably sensitive to the KIT and PDGFRA kinase inhibitors imatinib (also known as Gleevec) and sunitinib (Sutent), which have recently been approved as the standard therapeutic courses for patients with inoperable GIST. Sunitinib 134-140 platelet derived growth factor receptor alpha Homo sapiens 61-67 17208434-4 2007 Imatinib and sunitinib resistance generally result from secondary mutations in the KIT and/or PDGFRA kinase domains. Sunitinib 13-22 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 83-86 17208434-4 2007 Imatinib and sunitinib resistance generally result from secondary mutations in the KIT and/or PDGFRA kinase domains. Sunitinib 13-22 platelet derived growth factor receptor alpha Homo sapiens 94-100 17202116-1 2007 Sunitinib is an inhibitor of the vascular endothelial growth factor and platelet-derived growth factor receptors, and it has antitumor activity in metastatic renal cell carcinoma and gastrointestinal stromal tumors. Sunitinib 0-9 vascular endothelial growth factor A Homo sapiens 33-67 17296815-7 2007 Sunitinib is a multitargeted inhibitor of PDGFR, KIT, fms-like tyrosine kinase 3, and VEGFR. Sunitinib 0-9 platelet derived growth factor receptor beta Homo sapiens 42-47 17296815-7 2007 Sunitinib is a multitargeted inhibitor of PDGFR, KIT, fms-like tyrosine kinase 3, and VEGFR. Sunitinib 0-9 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 49-52 17296815-7 2007 Sunitinib is a multitargeted inhibitor of PDGFR, KIT, fms-like tyrosine kinase 3, and VEGFR. Sunitinib 0-9 fms related receptor tyrosine kinase 3 Homo sapiens 54-80 17296815-7 2007 Sunitinib is a multitargeted inhibitor of PDGFR, KIT, fms-like tyrosine kinase 3, and VEGFR. Sunitinib 0-9 kinase insert domain receptor Homo sapiens 86-91 17255305-5 2007 In contrast, two multitargeted receptor tyrosine kinase inhibitors that target both VEGF and PDGF receptors (sunitinib and AG013736) have shown > or =40% objective responses with clinically important duration. Sunitinib 109-118 vascular endothelial growth factor A Homo sapiens 84-88 17189392-4 2006 We review here the biology of RCC and how a combination of proximal and distal block of VHL/hypoxia-inducible factor alpha pathway by novel targeted agents, including sunitinib, sorafenib, bevacizumab, everolimus, and temsirolimus, has led to significant improvements in progression-free survival. Sunitinib 167-176 von Hippel-Lindau tumor suppressor Homo sapiens 88-91 17607922-8 2007 Sunitinib, another tyrosine kinase inhibitor, seems to be useful especially in patients with exon 9 mutations of c-kit, who usually have a worse response to imatinib. Sunitinib 0-9 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 113-118 16995874-5 2006 Interestingly, when ZD6474 was combined with sunitinib (SU11248; Sutent, Pfizer, Kalamazoo, MI, USA), a class III and V receptor tyrosine kinase inhibitor, the ZD6474-mediated growth inhibition was potentiated in association with further down-regulation of the mTOR targets p-p70S6K and p-4E-BP-1. Sunitinib 45-54 mechanistic target of rapamycin kinase Homo sapiens 261-265 16995874-5 2006 Interestingly, when ZD6474 was combined with sunitinib (SU11248; Sutent, Pfizer, Kalamazoo, MI, USA), a class III and V receptor tyrosine kinase inhibitor, the ZD6474-mediated growth inhibition was potentiated in association with further down-regulation of the mTOR targets p-p70S6K and p-4E-BP-1. Sunitinib 45-54 ribosomal protein S6 kinase B1 Homo sapiens 276-282 16860997-3 2006 Sunitinib, sorafenib and axitinib are new agents which belong to a class of drugs called kinase inhibitors and inhibit the VEGF, platelet-derived growth factor (PDGF) and c-KIT receptor tyrosine kinases. Sunitinib 0-9 vascular endothelial growth factor A Homo sapiens 123-127 16990784-0 2006 FLT3 K663Q is a novel AML-associated oncogenic kinase: Determination of biochemical properties and sensitivity to Sunitinib (SU11248). Sunitinib 114-123 FMS-like tyrosine kinase 3 Mus musculus 0-4 17064223-6 2006 So far, three anti-VEGF inhibitors, bevacizumab, sunitinib and sorafenib, have been approved for the treatment of solid human malignancies including colorectal cancer, gastrointestinal stromal tumours and renal cell carcinoma. Sunitinib 49-58 vascular endothelial growth factor A Homo sapiens 19-23 16990784-0 2006 FLT3 K663Q is a novel AML-associated oncogenic kinase: Determination of biochemical properties and sensitivity to Sunitinib (SU11248). Sunitinib 125-132 FMS-like tyrosine kinase 3 Mus musculus 0-4 16990784-8 2006 Of note, this mutation was potently inhibited by Sunitinib (SU11248), a previously described FLT3 kinase inhibitor. Sunitinib 49-58 FMS-like tyrosine kinase 3 Mus musculus 93-97 16990784-8 2006 Of note, this mutation was potently inhibited by Sunitinib (SU11248), a previously described FLT3 kinase inhibitor. Sunitinib 60-67 FMS-like tyrosine kinase 3 Mus musculus 93-97 16990784-9 2006 Sunitinib reduced the proliferation and induced apoptosis of transformed Ba/F3 cells expressing FLT3 K663Q. Sunitinib 0-9 FMS-like tyrosine kinase 3 Mus musculus 96-100 16990784-10 2006 The potency of Sunitinib against FLT3 K663Q was similar to its potency against FLT3 ITD mutations. Sunitinib 15-24 FMS-like tyrosine kinase 3 Mus musculus 33-37 17041096-2 2006 Thymidine uptake studies showed that sunitinib was active against EOL-1, MV4-11, and Kasumi-1 cells, which possessed activating mutations of the PDGFRalpha, FLT-3, and c-KIT genes, respectively, with IC(50)s of <30 nmol/L. Sunitinib 37-46 platelet derived growth factor receptor alpha Homo sapiens 145-155 16849418-0 2006 An orally administered multitarget tyrosine kinase inhibitor, SU11248, is a novel potent inhibitor of thyroid oncogenic RET/papillary thyroid cancer kinases. Sunitinib 62-69 ret proto-oncogene Homo sapiens 120-123 16849418-3 2006 OBJECTIVE: The objective of the study was to evaluate the efficacies of the recently developed kinase inhibitors SU11248, SU5416, and SU6668 in inhibition of RET/PTC. Sunitinib 113-120 ret proto-oncogene Homo sapiens 158-161 16849418-3 2006 OBJECTIVE: The objective of the study was to evaluate the efficacies of the recently developed kinase inhibitors SU11248, SU5416, and SU6668 in inhibition of RET/PTC. Sunitinib 113-120 ret proto-oncogene Homo sapiens 162-165 16849418-4 2006 DESIGN: SU11248, SU5416, and SU6668 were synthesized, and their inhibitory potencies were evaluated using an in vitro RET/PTC kinase assay. Sunitinib 8-15 ret proto-oncogene Homo sapiens 122-125 16849418-6 2006 RESULTS: An in vitro kinase assay revealed that SU5416, SU6668, and SU11248 inhibited phosphorylation of the synthetic tyrosine kinase substrate peptide E4Y by RET/PTC3 in a dose-dependent manner with IC(50) of approximately 944 nm for SU5416, 562 nm for SU6668, and 224 nm for SU11248. Sunitinib 68-75 ret proto-oncogene Homo sapiens 160-163 16849418-6 2006 RESULTS: An in vitro kinase assay revealed that SU5416, SU6668, and SU11248 inhibited phosphorylation of the synthetic tyrosine kinase substrate peptide E4Y by RET/PTC3 in a dose-dependent manner with IC(50) of approximately 944 nm for SU5416, 562 nm for SU6668, and 224 nm for SU11248. Sunitinib 68-75 nuclear receptor coactivator 4 Homo sapiens 164-168 16849418-6 2006 RESULTS: An in vitro kinase assay revealed that SU5416, SU6668, and SU11248 inhibited phosphorylation of the synthetic tyrosine kinase substrate peptide E4Y by RET/PTC3 in a dose-dependent manner with IC(50) of approximately 944 nm for SU5416, 562 nm for SU6668, and 224 nm for SU11248. Sunitinib 278-285 ret proto-oncogene Homo sapiens 160-163 16849418-7 2006 Thus, SU11248 effectively inhibits the kinase activity of RET/PTC3. Sunitinib 6-13 ret proto-oncogene Homo sapiens 58-61 16849418-7 2006 Thus, SU11248 effectively inhibits the kinase activity of RET/PTC3. Sunitinib 6-13 nuclear receptor coactivator 4 Homo sapiens 62-66 16849418-8 2006 RET/PTC-mediated Y705 phosphorylation of STAT3 was inhibited by addition of SU11248, and the inhibitory effects of SU11248 on the tyrosine phosphorylation and transcriptional activation of STAT3 were very closely correlated with decreased autophosphorylation of RET/PTC. Sunitinib 76-83 ret proto-oncogene Homo sapiens 0-3 16849418-8 2006 RET/PTC-mediated Y705 phosphorylation of STAT3 was inhibited by addition of SU11248, and the inhibitory effects of SU11248 on the tyrosine phosphorylation and transcriptional activation of STAT3 were very closely correlated with decreased autophosphorylation of RET/PTC. Sunitinib 76-83 ret proto-oncogene Homo sapiens 4-7 16849418-8 2006 RET/PTC-mediated Y705 phosphorylation of STAT3 was inhibited by addition of SU11248, and the inhibitory effects of SU11248 on the tyrosine phosphorylation and transcriptional activation of STAT3 were very closely correlated with decreased autophosphorylation of RET/PTC. Sunitinib 76-83 signal transducer and activator of transcription 3 Homo sapiens 41-46 16849418-8 2006 RET/PTC-mediated Y705 phosphorylation of STAT3 was inhibited by addition of SU11248, and the inhibitory effects of SU11248 on the tyrosine phosphorylation and transcriptional activation of STAT3 were very closely correlated with decreased autophosphorylation of RET/PTC. Sunitinib 115-122 ret proto-oncogene Homo sapiens 0-3 16849418-8 2006 RET/PTC-mediated Y705 phosphorylation of STAT3 was inhibited by addition of SU11248, and the inhibitory effects of SU11248 on the tyrosine phosphorylation and transcriptional activation of STAT3 were very closely correlated with decreased autophosphorylation of RET/PTC. Sunitinib 115-122 ret proto-oncogene Homo sapiens 4-7 16849418-8 2006 RET/PTC-mediated Y705 phosphorylation of STAT3 was inhibited by addition of SU11248, and the inhibitory effects of SU11248 on the tyrosine phosphorylation and transcriptional activation of STAT3 were very closely correlated with decreased autophosphorylation of RET/PTC. Sunitinib 115-122 signal transducer and activator of transcription 3 Homo sapiens 189-194 16849418-8 2006 RET/PTC-mediated Y705 phosphorylation of STAT3 was inhibited by addition of SU11248, and the inhibitory effects of SU11248 on the tyrosine phosphorylation and transcriptional activation of STAT3 were very closely correlated with decreased autophosphorylation of RET/PTC. Sunitinib 115-122 ret proto-oncogene Homo sapiens 262-265 16849418-8 2006 RET/PTC-mediated Y705 phosphorylation of STAT3 was inhibited by addition of SU11248, and the inhibitory effects of SU11248 on the tyrosine phosphorylation and transcriptional activation of STAT3 were very closely correlated with decreased autophosphorylation of RET/PTC. Sunitinib 115-122 ret proto-oncogene Homo sapiens 266-269 16849418-9 2006 SU11248 caused a complete morphological reversion of transformed NIH-RET/PTC3 cells and inhibited the growth of TPC-1 cells that have an endogenous RET/PTC1. Sunitinib 0-7 ret proto-oncogene Homo sapiens 69-72 16849418-9 2006 SU11248 caused a complete morphological reversion of transformed NIH-RET/PTC3 cells and inhibited the growth of TPC-1 cells that have an endogenous RET/PTC1. Sunitinib 0-7 nuclear receptor coactivator 4 Homo sapiens 73-77 16849418-9 2006 SU11248 caused a complete morphological reversion of transformed NIH-RET/PTC3 cells and inhibited the growth of TPC-1 cells that have an endogenous RET/PTC1. Sunitinib 0-7 ret proto-oncogene Homo sapiens 148-151 16849418-9 2006 SU11248 caused a complete morphological reversion of transformed NIH-RET/PTC3 cells and inhibited the growth of TPC-1 cells that have an endogenous RET/PTC1. Sunitinib 0-7 patched 1 Homo sapiens 152-156 16849418-10 2006 CONCLUSION: SU11248 is a highly effective tyrosine kinase inhibitor of the RET/PTC oncogenic kinase. Sunitinib 12-19 ret proto-oncogene Homo sapiens 75-78 16849418-10 2006 CONCLUSION: SU11248 is a highly effective tyrosine kinase inhibitor of the RET/PTC oncogenic kinase. Sunitinib 12-19 ret proto-oncogene Homo sapiens 79-82 17041096-2 2006 Thymidine uptake studies showed that sunitinib was active against EOL-1, MV4-11, and Kasumi-1 cells, which possessed activating mutations of the PDGFRalpha, FLT-3, and c-KIT genes, respectively, with IC(50)s of <30 nmol/L. Sunitinib 37-46 fms related receptor tyrosine kinase 3 Homo sapiens 157-162 17041096-2 2006 Thymidine uptake studies showed that sunitinib was active against EOL-1, MV4-11, and Kasumi-1 cells, which possessed activating mutations of the PDGFRalpha, FLT-3, and c-KIT genes, respectively, with IC(50)s of <30 nmol/L. Sunitinib 37-46 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 168-173 17041096-3 2006 In addition, sunitinib inhibited the proliferation of freshly isolated leukemia cells from patients possessing mutations in FLT3 gene. Sunitinib 13-22 fms related receptor tyrosine kinase 3 Homo sapiens 124-128 17041096-4 2006 Annexin V staining showed that sunitinib induced apoptosis of these cells. Sunitinib 31-40 annexin A5 Homo sapiens 0-9 17041096-5 2006 Sunitinib inhibited phosphorylation of FLT3 and PDGFRalpha in conjunction with blockade of mammalian target of rapamycin signaling in MV4-11 and EOL-1 cells, respectively. Sunitinib 0-9 fms related receptor tyrosine kinase 3 Homo sapiens 39-43 17041096-5 2006 Sunitinib inhibited phosphorylation of FLT3 and PDGFRalpha in conjunction with blockade of mammalian target of rapamycin signaling in MV4-11 and EOL-1 cells, respectively. Sunitinib 0-9 platelet derived growth factor receptor alpha Homo sapiens 48-58 16974118-8 2006 This opens interesting new treatment strategies including blockade of VEGF with the monoclonal antibody bevacizumab (Avastin) and inhibition of VEGF receptor tyrosine kinases with small oral molecules such as sunitinib (SU11248, Sutent) or PTK787. Sunitinib 209-218 vascular endothelial growth factor A Homo sapiens 144-148 16916320-0 2006 Effect of SU11248 on gastrointestinal stromal tumor-T1 cells: enhancement of growth inhibition via inhibition of 3-kinase/Akt/mammalian target of rapamycin signaling. Sunitinib 10-17 AKT serine/threonine kinase 1 Homo sapiens 122-125 16916320-0 2006 Effect of SU11248 on gastrointestinal stromal tumor-T1 cells: enhancement of growth inhibition via inhibition of 3-kinase/Akt/mammalian target of rapamycin signaling. Sunitinib 10-17 mechanistic target of rapamycin kinase Homo sapiens 126-155 16916320-3 2006 Taking advantage of GIST-T1 cells, which possess an activating mutation in exon 11 of the c-KIT gene, we examined the medicinal action of SU11248 in GIST cells. Sunitinib 138-145 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 90-95 16916320-5 2006 SU11248 (10 or 20 nM, 48 h) activated caspase-3 and induced apoptosis of GIST-T1 cells as measured by caspase assay, annexin V staining and cleavage of poly (ADP-ribose) polymerase. Sunitinib 0-7 caspase 3 Homo sapiens 38-47 16916320-5 2006 SU11248 (10 or 20 nM, 48 h) activated caspase-3 and induced apoptosis of GIST-T1 cells as measured by caspase assay, annexin V staining and cleavage of poly (ADP-ribose) polymerase. Sunitinib 0-7 annexin A5 Homo sapiens 117-126 16916320-5 2006 SU11248 (10 or 20 nM, 48 h) activated caspase-3 and induced apoptosis of GIST-T1 cells as measured by caspase assay, annexin V staining and cleavage of poly (ADP-ribose) polymerase. Sunitinib 0-7 poly(ADP-ribose) polymerase 1 Homo sapiens 152-180 16916320-6 2006 Western blot analyses found that SU11248 blocked autophosphorylation of c-KIT in association with inhibition of its downstream effectors, including Akt and extracellular signal-regulated kinase, but not signal transducers and activators of transcription. Sunitinib 33-40 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 72-77 16916320-6 2006 Western blot analyses found that SU11248 blocked autophosphorylation of c-KIT in association with inhibition of its downstream effectors, including Akt and extracellular signal-regulated kinase, but not signal transducers and activators of transcription. Sunitinib 33-40 AKT serine/threonine kinase 1 Homo sapiens 148-151 16916320-7 2006 Interestingly, when phosphatidylinositol 3-kinase/Akt/mammalian target of rapamycin signaling was blocked simultaneously by either LY294002 or rapamycin, growth inhibition mediated by SU11248 was potentiated. Sunitinib 184-191 AKT serine/threonine kinase 1 Homo sapiens 50-53 16916320-7 2006 Interestingly, when phosphatidylinositol 3-kinase/Akt/mammalian target of rapamycin signaling was blocked simultaneously by either LY294002 or rapamycin, growth inhibition mediated by SU11248 was potentiated. Sunitinib 184-191 mechanistic target of rapamycin kinase Homo sapiens 54-83 16685460-3 2006 Bevacizumab (Avastin), Sunitinib (Sutent) and Sorafenib (Nexavar) are anti-cancer drugs targeted to VEGF signaling pathway. Sunitinib 23-32 vascular endothelial growth factor A Homo sapiens 100-104 16859583-1 2006 Sunitinib and sorafenib are multitargeted receptor tyrosine kinase inhibitors of the vascular endothelial growth factor and platelet-derived growth factor receptor families with antiangiogenic and antitumor activity in metastatic renal cell carcinoma. Sunitinib 0-9 vascular endothelial growth factor A Homo sapiens 85-119 16910860-0 2006 A novel vascular endothelial growth factor receptor 2 inhibitor, SU11248, suppresses choroidal neovascularization in vivo. Sunitinib 65-72 kinase insert domain protein receptor Mus musculus 8-53 16910860-1 2006 AIM: An oral, multitargeted receptor tyrosine kinase inhibitor, SU11248, inhibits vascular endothelial growth factor receptor 2 (VEG FR2, kinase domain receptor F1K1), platelet-derived growth factor receptor, stem cell factor receptor (Kit), and fetal liver tyrosine kinase 3. Sunitinib 64-71 kinase insert domain protein receptor Mus musculus 82-127 16910860-1 2006 AIM: An oral, multitargeted receptor tyrosine kinase inhibitor, SU11248, inhibits vascular endothelial growth factor receptor 2 (VEG FR2, kinase domain receptor F1K1), platelet-derived growth factor receptor, stem cell factor receptor (Kit), and fetal liver tyrosine kinase 3. Sunitinib 64-71 kinase insert domain protein receptor Mus musculus 129-136 16910860-1 2006 AIM: An oral, multitargeted receptor tyrosine kinase inhibitor, SU11248, inhibits vascular endothelial growth factor receptor 2 (VEG FR2, kinase domain receptor F1K1), platelet-derived growth factor receptor, stem cell factor receptor (Kit), and fetal liver tyrosine kinase 3. Sunitinib 64-71 receptor-like tyrosine kinase Mus musculus 103-125 16850127-3 2006 To date, the vascular endothelial growth factor receptor (VEGFR) pathway has been the most promising target, and two agents (BAY 43-9006 and SU 11248) that inhibit not only VEGFR but also other receptors, including platelet-derived growth factor receptor (PDGFR), FMS-like tyrosine kinase 3 (FLT3), KIT, and Raf kinase, were recently approved by the FDA for advanced RCC. Sunitinib 141-149 kinase insert domain receptor Homo sapiens 173-178 16850127-3 2006 To date, the vascular endothelial growth factor receptor (VEGFR) pathway has been the most promising target, and two agents (BAY 43-9006 and SU 11248) that inhibit not only VEGFR but also other receptors, including platelet-derived growth factor receptor (PDGFR), FMS-like tyrosine kinase 3 (FLT3), KIT, and Raf kinase, were recently approved by the FDA for advanced RCC. Sunitinib 141-149 platelet derived growth factor receptor beta Homo sapiens 215-254 16850127-3 2006 To date, the vascular endothelial growth factor receptor (VEGFR) pathway has been the most promising target, and two agents (BAY 43-9006 and SU 11248) that inhibit not only VEGFR but also other receptors, including platelet-derived growth factor receptor (PDGFR), FMS-like tyrosine kinase 3 (FLT3), KIT, and Raf kinase, were recently approved by the FDA for advanced RCC. Sunitinib 141-149 platelet derived growth factor receptor beta Homo sapiens 256-261 16850127-3 2006 To date, the vascular endothelial growth factor receptor (VEGFR) pathway has been the most promising target, and two agents (BAY 43-9006 and SU 11248) that inhibit not only VEGFR but also other receptors, including platelet-derived growth factor receptor (PDGFR), FMS-like tyrosine kinase 3 (FLT3), KIT, and Raf kinase, were recently approved by the FDA for advanced RCC. Sunitinib 141-149 fms related receptor tyrosine kinase 3 Homo sapiens 264-290 16850127-3 2006 To date, the vascular endothelial growth factor receptor (VEGFR) pathway has been the most promising target, and two agents (BAY 43-9006 and SU 11248) that inhibit not only VEGFR but also other receptors, including platelet-derived growth factor receptor (PDGFR), FMS-like tyrosine kinase 3 (FLT3), KIT, and Raf kinase, were recently approved by the FDA for advanced RCC. Sunitinib 141-149 fms related receptor tyrosine kinase 3 Homo sapiens 292-296 16850127-3 2006 To date, the vascular endothelial growth factor receptor (VEGFR) pathway has been the most promising target, and two agents (BAY 43-9006 and SU 11248) that inhibit not only VEGFR but also other receptors, including platelet-derived growth factor receptor (PDGFR), FMS-like tyrosine kinase 3 (FLT3), KIT, and Raf kinase, were recently approved by the FDA for advanced RCC. Sunitinib 141-149 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 299-302 16685460-3 2006 Bevacizumab (Avastin), Sunitinib (Sutent) and Sorafenib (Nexavar) are anti-cancer drugs targeted to VEGF signaling pathway. Sunitinib 34-40 vascular endothelial growth factor A Homo sapiens 100-104 16731761-2 2006 SU11248, a small-molecule inhibitor targeting class III/V receptor tyrosine kinases, including the platelet-derived growth factor (PDGF) and vascular endothelial growth factor (VEGF) receptors, KIT and FLT3, exhibits direct effects on cancer cells as well as antiangiogenic activity. Sunitinib 0-7 vascular endothelial growth factor A Homo sapiens 141-175 16731761-5 2006 To elucidate the specific contributions of inhibition of PDGF and VEGF receptors to the in vivo efficacy of SU11248, we employed two selective inhibitors, SU10944 (VEGF receptor inhibitor) and Gleevec (PDGF receptor inhibitor). Sunitinib 108-115 vascular endothelial growth factor A Homo sapiens 66-70 16731761-9 2006 The one exception was a model driven by an activated mutant of FLT3, in which the activity of SU11248, which targets FLT3, was greater than that of SU10944 plus Gleevec. Sunitinib 94-101 fms related receptor tyrosine kinase 3 Homo sapiens 63-67 16731761-9 2006 The one exception was a model driven by an activated mutant of FLT3, in which the activity of SU11248, which targets FLT3, was greater than that of SU10944 plus Gleevec. Sunitinib 94-101 fms related receptor tyrosine kinase 3 Homo sapiens 117-121 16638875-10 2006 In agreement with these results, two of the three imatinib-resistant patients with the KIT-V654A mutation responded to SU11248 treatment. Sunitinib 119-126 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 87-90 16638875-11 2006 CONCLUSIONS: These studies suggest that SU11248 may be a useful therapeutic agent to treat gastrointestinal stromal tumors harboring the imatinib-resistant KIT-V654A or KIT-T670I mutations, but it has no effect on the activity of the PDGFRA-D842V mutant. Sunitinib 40-47 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 156-159 16638875-4 2006 We tested the activity of SU11248, an orally active small-molecule tyrosine kinase inhibitor, to inhibit important imatinib-resistant KIT and PDGFRA mutants. Sunitinib 26-33 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 134-137 16638875-11 2006 CONCLUSIONS: These studies suggest that SU11248 may be a useful therapeutic agent to treat gastrointestinal stromal tumors harboring the imatinib-resistant KIT-V654A or KIT-T670I mutations, but it has no effect on the activity of the PDGFRA-D842V mutant. Sunitinib 40-47 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 169-172 16638875-4 2006 We tested the activity of SU11248, an orally active small-molecule tyrosine kinase inhibitor, to inhibit important imatinib-resistant KIT and PDGFRA mutants. Sunitinib 26-33 platelet derived growth factor receptor alpha Homo sapiens 142-148 16638875-6 2006 Three patients with the KIT-V654A mutation were treated with SU11248. Sunitinib 61-68 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 24-27 16638875-11 2006 CONCLUSIONS: These studies suggest that SU11248 may be a useful therapeutic agent to treat gastrointestinal stromal tumors harboring the imatinib-resistant KIT-V654A or KIT-T670I mutations, but it has no effect on the activity of the PDGFRA-D842V mutant. Sunitinib 40-47 platelet derived growth factor receptor alpha Homo sapiens 234-240 16638875-7 2006 RESULTS: Based on ex vivo assays, SU11248 potently inhibits KIT kinase activity of V654A and T670I mutants and suppresses proliferation of the cells expressing these mutations. Sunitinib 34-41 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 60-63 16638875-8 2006 Sensitivity of KIT-V654A and KIT-T670I mutants to SU11248 was confirmed using cell lines expressing these mutants. Sunitinib 50-57 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 15-18 16638875-8 2006 Sensitivity of KIT-V654A and KIT-T670I mutants to SU11248 was confirmed using cell lines expressing these mutants. Sunitinib 50-57 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 29-32 16556328-3 2006 For example Imatinib/Gleevec (primarily a bcr/abl kinase inhibitor) or SU11248 (mainly a VEGFR inhibitor) are also potent inhibitors of PDGFR and other kinases. Sunitinib 71-78 kinase insert domain receptor Homo sapiens 89-94 16648572-6 2006 SU11248 is a potent inhibitor of CSF-1R in the enzyme assay (IC50 = 7 nmol/L) and inhibits receptor phosphorylation in the cellular assay (IC50 = 61 nmol/L). Sunitinib 0-7 colony stimulating factor 1 receptor Homo sapiens 33-39 16556328-3 2006 For example Imatinib/Gleevec (primarily a bcr/abl kinase inhibitor) or SU11248 (mainly a VEGFR inhibitor) are also potent inhibitors of PDGFR and other kinases. Sunitinib 71-78 platelet derived growth factor receptor beta Homo sapiens 136-141 16425985-2 2005 SU11248 and AG013736 are small-molecule inhibitors of the tyrosine kinase portion of the VEGF and PDGF receptors. Sunitinib 0-7 vascular endothelial growth factor A Homo sapiens 89-93 16640800-2 2006 These include the small-molecule receptor tyrosine kinase (TK) inhibitors ZD6474, sorafenib, sunitinib malate, and AG-013736, all of which inhibit VEGF receptor TK activity. Sunitinib 93-109 vascular endothelial growth factor A Homo sapiens 147-151 18393778-6 2006 In addition, other KIT tyrosine kinase inhibitors with anti-VEGF receptor inhibitory activity, such as SU11248, PTK787/ZK787 and AMG 706, are currently being explored as second line monotherapy for imatinib mesylate-resistant GIST. Sunitinib 103-110 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 19-22 16330672-2 2006 SU11248 (sunitinib malate), a small molecule inhibitor with high binding affinity for VEGF and PDGF receptors, was tested for clinical activity in patients with metastatic RCC. Sunitinib 0-7 vascular endothelial growth factor A Homo sapiens 86-90 16330672-9 2006 CONCLUSION: SU11248, a multitargeted receptor tyrosine kinase inhibitor of VEGF and PDGF receptors, demonstrates antitumor activity in metastatic RCC as second-line therapy, a setting where no effective systemic therapy is presently recognized. Sunitinib 12-19 vascular endothelial growth factor A Homo sapiens 75-79 16425993-2 2005 Significant advances in the treatment of clear-cell RCC have been derived from agents that target these pathways, including the multiple-kinase inhibitors (MKIs) sorafenib, sunitinib, and AG013736, which target multiple VEGFRs as well as PDGFR-beta. Sunitinib 173-182 kinase insert domain receptor Homo sapiens 220-226 15459012-1 2005 Fifteen patients with refractory AML were treated in a phase 1 study with SU11248, an oral kinase inhibitor of fms-like tyrosine kinase 3 (Flt3), Kit, vascular endothelial growth factor (VEGF), and platelet-derived growth factor (PDGF) receptors. Sunitinib 74-81 fms related receptor tyrosine kinase 3 Homo sapiens 111-137 16185167-4 2005 Results using small-molecule inhibitors of VEGF, FLT3, KIT and platelet-derived growth factor receptor tyrosine kinases, such as sunitinib, show a 40% objective response rate. Sunitinib 129-138 vascular endothelial growth factor A Homo sapiens 43-47 15557593-4 2005 MATERIALS AND METHODS: Seeking to improve efficacy against otherwise intractable end-stage pancreatic islet tumors, two receptor tyrosine kinase inhibitors, imatinib and SU11248, were used to disrupt PDGFR-mediated pericyte support of tumor endothelial cells in concert with maximum-tolerated dose (MTD) or metronomic chemotherapy and/or VEGFR inhibition. Sunitinib 170-177 platelet derived growth factor receptor, beta polypeptide Mus musculus 200-205 16115922-1 2005 PURPOSE: To compare two dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) techniques in terms of their ability in assessing the early antiangiogenic effect of SU11248, a novel selective multitargeted tyrosine kinase inhibitor, that exhibits direct antitumor and antiangiogenic activity via inhibition of the receptor tyrosine kinases platelet-derived growth factor receptor, vascular endothelial growth factor receptor, KIT, and FLT3. Sunitinib 172-179 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 433-436 16115922-1 2005 PURPOSE: To compare two dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) techniques in terms of their ability in assessing the early antiangiogenic effect of SU11248, a novel selective multitargeted tyrosine kinase inhibitor, that exhibits direct antitumor and antiangiogenic activity via inhibition of the receptor tyrosine kinases platelet-derived growth factor receptor, vascular endothelial growth factor receptor, KIT, and FLT3. Sunitinib 172-179 fms related receptor tyrosine kinase 3 Homo sapiens 442-446 16046538-6 2005 The KIT/FLT3 inhibitor SU-11248 potently inhibits the imatinib-resistant KIT(V559D/T670I) kinase, consistent with the clinical efficacy of SU-11248 against imatinib-resistant gastrointestinal tumors, and the EGFR inhibitors EKB-569 and CI-1033, but not GW-572016 and ZD-6474, potently inhibit the gefitinib- and erlotinib-resistant EGFR(L858R/T790M) kinase. Sunitinib 23-31 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 4-7 16046538-6 2005 The KIT/FLT3 inhibitor SU-11248 potently inhibits the imatinib-resistant KIT(V559D/T670I) kinase, consistent with the clinical efficacy of SU-11248 against imatinib-resistant gastrointestinal tumors, and the EGFR inhibitors EKB-569 and CI-1033, but not GW-572016 and ZD-6474, potently inhibit the gefitinib- and erlotinib-resistant EGFR(L858R/T790M) kinase. Sunitinib 23-31 fms related receptor tyrosine kinase 3 Homo sapiens 8-12 16046538-6 2005 The KIT/FLT3 inhibitor SU-11248 potently inhibits the imatinib-resistant KIT(V559D/T670I) kinase, consistent with the clinical efficacy of SU-11248 against imatinib-resistant gastrointestinal tumors, and the EGFR inhibitors EKB-569 and CI-1033, but not GW-572016 and ZD-6474, potently inhibit the gefitinib- and erlotinib-resistant EGFR(L858R/T790M) kinase. Sunitinib 23-31 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 73-76 16046538-6 2005 The KIT/FLT3 inhibitor SU-11248 potently inhibits the imatinib-resistant KIT(V559D/T670I) kinase, consistent with the clinical efficacy of SU-11248 against imatinib-resistant gastrointestinal tumors, and the EGFR inhibitors EKB-569 and CI-1033, but not GW-572016 and ZD-6474, potently inhibit the gefitinib- and erlotinib-resistant EGFR(L858R/T790M) kinase. Sunitinib 23-31 epidermal growth factor receptor Homo sapiens 208-212 16046538-6 2005 The KIT/FLT3 inhibitor SU-11248 potently inhibits the imatinib-resistant KIT(V559D/T670I) kinase, consistent with the clinical efficacy of SU-11248 against imatinib-resistant gastrointestinal tumors, and the EGFR inhibitors EKB-569 and CI-1033, but not GW-572016 and ZD-6474, potently inhibit the gefitinib- and erlotinib-resistant EGFR(L858R/T790M) kinase. Sunitinib 23-31 epidermal growth factor receptor Homo sapiens 332-336 16046538-6 2005 The KIT/FLT3 inhibitor SU-11248 potently inhibits the imatinib-resistant KIT(V559D/T670I) kinase, consistent with the clinical efficacy of SU-11248 against imatinib-resistant gastrointestinal tumors, and the EGFR inhibitors EKB-569 and CI-1033, but not GW-572016 and ZD-6474, potently inhibit the gefitinib- and erlotinib-resistant EGFR(L858R/T790M) kinase. Sunitinib 139-147 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 4-7 16046538-6 2005 The KIT/FLT3 inhibitor SU-11248 potently inhibits the imatinib-resistant KIT(V559D/T670I) kinase, consistent with the clinical efficacy of SU-11248 against imatinib-resistant gastrointestinal tumors, and the EGFR inhibitors EKB-569 and CI-1033, but not GW-572016 and ZD-6474, potently inhibit the gefitinib- and erlotinib-resistant EGFR(L858R/T790M) kinase. Sunitinib 139-147 fms related receptor tyrosine kinase 3 Homo sapiens 8-12 15304385-0 2004 Synergistic effect of SU11248 with cytarabine or daunorubicin on FLT3 ITD-positive leukemic cells. Sunitinib 22-29 fms related receptor tyrosine kinase 3 Homo sapiens 65-69 15304385-4 2004 SU11248 had additive-to-synergistic inhibitory effects on FLT3-dependent leukemic cell proliferation when combined with cytarabine or daunorubicin. Sunitinib 0-7 fms related receptor tyrosine kinase 3 Homo sapiens 58-62 14985702-8 2004 One such potential biomarker, cadherin-11, was further evaluated at the protein level and was found to have increased expression in xenograft tumors after SU11248 treatment. Sunitinib 155-162 cadherin 11 Homo sapiens 30-41 15304385-6 2004 SU11248 inhibited the proliferation of primary AML myeloblasts expressing mutant FLT3 ITD but not WT FLT3 protein. Sunitinib 0-7 fms related receptor tyrosine kinase 3 Homo sapiens 81-85 15304385-7 2004 Combining SU11248 and cytarabine synergistically inhibited the proliferation of primary AML myeloblasts expressing FLT3 ITD but not WT FLT3 protein. Sunitinib 10-17 fms related receptor tyrosine kinase 3 Homo sapiens 115-119 15304385-8 2004 These data suggest that the addition of potent FLT3 inhibitors such as SU11248 to AML chemotherapy regimens could result in improved treatment results. Sunitinib 71-78 fms related receptor tyrosine kinase 3 Homo sapiens 47-51 15648955-1 2004 SUGEN is developing SU-11248, an orally active inhibitor of platelet-derived growth factor tyrosine kinase (TK) and other TK signaling pathways, as a potential anticancer agent. Sunitinib 20-28 TXK tyrosine kinase Homo sapiens 91-106 15648955-1 2004 SUGEN is developing SU-11248, an orally active inhibitor of platelet-derived growth factor tyrosine kinase (TK) and other TK signaling pathways, as a potential anticancer agent. Sunitinib 20-28 TXK tyrosine kinase Homo sapiens 108-110 15648955-1 2004 SUGEN is developing SU-11248, an orally active inhibitor of platelet-derived growth factor tyrosine kinase (TK) and other TK signaling pathways, as a potential anticancer agent. Sunitinib 20-28 TXK tyrosine kinase Homo sapiens 122-124 14985702-10 2004 Importantly, SU11248 treatment also resulted in increased expression of cadherin-11 protein in human tumor biopsies in three out of seven patients examined and confirms the feasibility of using transcriptional profiling of preclinical models to identify clinically translatable biomarkers. Sunitinib 13-20 cadherin 11 Homo sapiens 72-83 14753710-1 2004 SU11248 is a potent inhibitor of PDGFR, VEGFR, KIT, and Flt3, and is currently under Phase I clinical evaluation as an anticancer drug. Sunitinib 0-7 platelet derived growth factor receptor beta Homo sapiens 33-38 14753710-1 2004 SU11248 is a potent inhibitor of PDGFR, VEGFR, KIT, and Flt3, and is currently under Phase I clinical evaluation as an anticancer drug. Sunitinib 0-7 kinase insert domain receptor Homo sapiens 40-45 14753710-1 2004 SU11248 is a potent inhibitor of PDGFR, VEGFR, KIT, and Flt3, and is currently under Phase I clinical evaluation as an anticancer drug. Sunitinib 0-7 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 47-50 14753710-1 2004 SU11248 is a potent inhibitor of PDGFR, VEGFR, KIT, and Flt3, and is currently under Phase I clinical evaluation as an anticancer drug. Sunitinib 0-7 fms related receptor tyrosine kinase 3 Homo sapiens 56-60 15609077-0 2004 The receptor tyrosine kinase inhibitor SU11248 impedes endothelial cell migration, tubule formation, and blood vessel formation in vivo, but has little effect on existing tumor vessels. Sunitinib 39-46 ret proto-oncogene Homo sapiens 4-28 15609077-2 2004 To determine whether the vascular endothelial growth factor (VEGF) receptor is a molecular target to prevent metastatic disease, we utilized a non-specific inhibitor of the VEGF receptor, SU11248. Sunitinib 188-195 vascular endothelial growth factor A Homo sapiens 61-65 15609077-2 2004 To determine whether the vascular endothelial growth factor (VEGF) receptor is a molecular target to prevent metastatic disease, we utilized a non-specific inhibitor of the VEGF receptor, SU11248. Sunitinib 188-195 vascular endothelial growth factor A Homo sapiens 173-177 15000426-2 2004 SU011248 is an oral, multitargeted receptor tyrosine kinase inhibitor (anti PDGFR, VEGFR, Kit, and Flt3) for the treatment of solid tumors. Sunitinib 0-8 platelet derived growth factor receptor beta Homo sapiens 76-81 15000426-2 2004 SU011248 is an oral, multitargeted receptor tyrosine kinase inhibitor (anti PDGFR, VEGFR, Kit, and Flt3) for the treatment of solid tumors. Sunitinib 0-8 kinase insert domain receptor Homo sapiens 83-88 15000426-2 2004 SU011248 is an oral, multitargeted receptor tyrosine kinase inhibitor (anti PDGFR, VEGFR, Kit, and Flt3) for the treatment of solid tumors. Sunitinib 0-8 fms related receptor tyrosine kinase 3 Homo sapiens 99-103 12748309-4 2003 The biological significance of KIT inhibition was evaluated in vivo by treating mice bearing s.c. NCI-H526 tumors with SU11248 or another structurally unrelated KIT inhibitor, STI571 (Gleevec), which is also known to inhibit Bcr-Abl and PDGFRbeta. Sunitinib 119-126 KIT proto-oncogene receptor tyrosine kinase Mus musculus 31-34 14654525-0 2003 An innovative phase I clinical study demonstrates inhibition of FLT3 phosphorylation by SU11248 in acute myeloid leukemia patients. Sunitinib 88-95 fms related receptor tyrosine kinase 3 Homo sapiens 64-68 14654525-2 2003 SU11248 is an oral multitargeted kinase inhibitor with selectivity for fms-related tyrosine kinase 3/Flk2 (FLT3), platelet-derived growth factor receptor alpha/beta, vascular endothelial growth factor receptor 1/2, and KIT receptor tyrosine kinases. Sunitinib 0-7 fms related receptor tyrosine kinase 3 Homo sapiens 71-100 14654525-2 2003 SU11248 is an oral multitargeted kinase inhibitor with selectivity for fms-related tyrosine kinase 3/Flk2 (FLT3), platelet-derived growth factor receptor alpha/beta, vascular endothelial growth factor receptor 1/2, and KIT receptor tyrosine kinases. Sunitinib 0-7 fms related receptor tyrosine kinase 3 Homo sapiens 101-105 14654525-2 2003 SU11248 is an oral multitargeted kinase inhibitor with selectivity for fms-related tyrosine kinase 3/Flk2 (FLT3), platelet-derived growth factor receptor alpha/beta, vascular endothelial growth factor receptor 1/2, and KIT receptor tyrosine kinases. Sunitinib 0-7 fms related receptor tyrosine kinase 3 Homo sapiens 107-111 14654525-2 2003 SU11248 is an oral multitargeted kinase inhibitor with selectivity for fms-related tyrosine kinase 3/Flk2 (FLT3), platelet-derived growth factor receptor alpha/beta, vascular endothelial growth factor receptor 1/2, and KIT receptor tyrosine kinases. Sunitinib 0-7 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 219-222 14654525-4 2003 We conducted an innovative single-dose clinical study with a primary objective to demonstrate inhibition of FLT3 phosphorylation by SU11248 in AML. Sunitinib 132-139 fms related receptor tyrosine kinase 3 Homo sapiens 108-112 14578466-1 2003 SU11248 is an oral multitargeted tyrosine kinase inhibitor with antitumor and antiangiogenic activities through targeting platelet-derived growth factor receptor, vascular endothelial growth factor receptor, KIT, and FLT3, the first three of which are expressed in human breast cancer and/or its supporting tissues. Sunitinib 0-7 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 208-211 14578466-1 2003 SU11248 is an oral multitargeted tyrosine kinase inhibitor with antitumor and antiangiogenic activities through targeting platelet-derived growth factor receptor, vascular endothelial growth factor receptor, KIT, and FLT3, the first three of which are expressed in human breast cancer and/or its supporting tissues. Sunitinib 0-7 fms related receptor tyrosine kinase 3 Homo sapiens 217-221 14578466-3 2003 SU11248 was administered as a monotherapy to (1) mouse mammary tumor virus-v-Ha-ras mice and 7,12-dimethylbenz(a)anthracene-treated rats bearing mammary tumors and (2) mice bearing human breast cancer xenografts of s.c. MX-1 tumors and osseous metastasis of a MDA-MB-435-derived cell line (435/HAL-Luc). Sunitinib 0-7 histidine ammonia-lyase Homo sapiens 294-297 14578466-7 2003 SU11248 treatment in combination with docetaxel effectively prolonged survival of mice, with 435/HAL-Luc cancer xenografts established in bone compared with either agent alone (P < 0.05). Sunitinib 0-7 histidine ammonia lyase Mus musculus 97-100 12748309-0 2003 SU11248 inhibits KIT and platelet-derived growth factor receptor beta in preclinical models of human small cell lung cancer. Sunitinib 0-7 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 17-20 12748309-0 2003 SU11248 inhibits KIT and platelet-derived growth factor receptor beta in preclinical models of human small cell lung cancer. Sunitinib 0-7 platelet derived growth factor receptor beta Homo sapiens 25-69 12748309-1 2003 The purpose of this study was to evaluate the activity of the indolinone kinase inhibitor SU11248 against the receptor tyrosine kinase KIT in vitro and in vivo, examine the role of KIT in small cell lung cancer (SCLC), and anticipate clinical utility of SU11248 in SCLC. Sunitinib 90-97 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 135-138 12748309-2 2003 SU11248 is an oral, multitargeted tyrosine kinase inhibitor with direct antitumor and antiangiogenic activity through targeting platelet-derived growth factor receptor (PDGFR), vascular endothelial growth factor receptor, KIT, and FLT3 receptors. Sunitinib 0-7 platelet derived growth factor receptor beta Homo sapiens 128-167 12748309-2 2003 SU11248 is an oral, multitargeted tyrosine kinase inhibitor with direct antitumor and antiangiogenic activity through targeting platelet-derived growth factor receptor (PDGFR), vascular endothelial growth factor receptor, KIT, and FLT3 receptors. Sunitinib 0-7 platelet derived growth factor receptor beta Homo sapiens 169-174 12748309-2 2003 SU11248 is an oral, multitargeted tyrosine kinase inhibitor with direct antitumor and antiangiogenic activity through targeting platelet-derived growth factor receptor (PDGFR), vascular endothelial growth factor receptor, KIT, and FLT3 receptors. Sunitinib 0-7 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 222-225 12748309-2 2003 SU11248 is an oral, multitargeted tyrosine kinase inhibitor with direct antitumor and antiangiogenic activity through targeting platelet-derived growth factor receptor (PDGFR), vascular endothelial growth factor receptor, KIT, and FLT3 receptors. Sunitinib 0-7 fms related receptor tyrosine kinase 3 Homo sapiens 231-235 12748309-3 2003 Treatment of the KIT-expressing SCLC-derived NCI-H526 cell line in vitro with SU11248 resulted in dose-dependent inhibition of stem cell factor-stimulated KIT phosphotyrosine levels and proliferation. Sunitinib 78-85 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 17-20 12748309-3 2003 Treatment of the KIT-expressing SCLC-derived NCI-H526 cell line in vitro with SU11248 resulted in dose-dependent inhibition of stem cell factor-stimulated KIT phosphotyrosine levels and proliferation. Sunitinib 78-85 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 155-158 12873999-2 2003 The novel broad spectrum receptor tyrosine kinase inhibitor, SU11248, inhibits vascular endothelial growth factor receptor 2, platelet-derived growth factor receptor, c-kit, and fetal liver tyrosine kinase 3. Sunitinib 61-68 kinase insert domain protein receptor Mus musculus 79-124 12873999-2 2003 The novel broad spectrum receptor tyrosine kinase inhibitor, SU11248, inhibits vascular endothelial growth factor receptor 2, platelet-derived growth factor receptor, c-kit, and fetal liver tyrosine kinase 3. Sunitinib 61-68 KIT proto-oncogene receptor tyrosine kinase Mus musculus 167-172 12531805-0 2003 SU11248 is a novel FLT3 tyrosine kinase inhibitor with potent activity in vitro and in vivo. Sunitinib 0-7 fms related receptor tyrosine kinase 3 Homo sapiens 19-23 12531805-5 2003 SU11248 is a recently described selective inhibitor with selectivity for split kinase domain RTKs, including platelet-derived growth factor receptors, vascular endothelial growth factor receptors, and KIT. Sunitinib 0-7 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 151-204 12531805-6 2003 We show that SU11248 also has potent activity against wild-type FLT3 (FLT3-WT), FLT3-ITD, and FLT3 activation loop (FLT3-Asp835) mutants in phosphorylation assays. Sunitinib 13-20 fms related receptor tyrosine kinase 3 Homo sapiens 64-68 12531805-6 2003 We show that SU11248 also has potent activity against wild-type FLT3 (FLT3-WT), FLT3-ITD, and FLT3 activation loop (FLT3-Asp835) mutants in phosphorylation assays. Sunitinib 13-20 fms related receptor tyrosine kinase 3 Homo sapiens 70-77 12531805-6 2003 We show that SU11248 also has potent activity against wild-type FLT3 (FLT3-WT), FLT3-ITD, and FLT3 activation loop (FLT3-Asp835) mutants in phosphorylation assays. Sunitinib 13-20 fms related receptor tyrosine kinase 3 Homo sapiens 70-74 12531805-6 2003 We show that SU11248 also has potent activity against wild-type FLT3 (FLT3-WT), FLT3-ITD, and FLT3 activation loop (FLT3-Asp835) mutants in phosphorylation assays. Sunitinib 13-20 fms related receptor tyrosine kinase 3 Homo sapiens 70-74 12531805-6 2003 We show that SU11248 also has potent activity against wild-type FLT3 (FLT3-WT), FLT3-ITD, and FLT3 activation loop (FLT3-Asp835) mutants in phosphorylation assays. Sunitinib 13-20 fms related receptor tyrosine kinase 3 Homo sapiens 70-74 12531805-7 2003 SU11248 inhibits FLT3-driven phosphorylation and induces apoptosis in vitro. Sunitinib 0-7 fms related receptor tyrosine kinase 3 Homo sapiens 17-21 12531805-8 2003 In addition, SU11248 inhibits FLT3-induced VEGF production. Sunitinib 13-20 fms related receptor tyrosine kinase 3 Homo sapiens 30-34 12531805-8 2003 In addition, SU11248 inhibits FLT3-induced VEGF production. Sunitinib 13-20 vascular endothelial growth factor A Homo sapiens 43-47 12748309-7 2003 Likewise, phospho-PDGFRbeta levels contributed by tumor stroma and with known involvement in angiogenesis were strongly inhibited by SU11248 and less so by STI571. Sunitinib 133-140 platelet derived growth factor receptor beta Homo sapiens 18-27 12748309-9 2003 These results expand the profile of SU11248 as a KIT signaling inhibitor and suggest that SU11248 may have clinical potential in the treatment of SCLC via direct antitumor activity mediated via KIT as well as tumor angiogenesis via vascular endothelial growth factor receptor FLK1/KDR and PDGFRbeta. Sunitinib 36-43 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 49-52 12748309-9 2003 These results expand the profile of SU11248 as a KIT signaling inhibitor and suggest that SU11248 may have clinical potential in the treatment of SCLC via direct antitumor activity mediated via KIT as well as tumor angiogenesis via vascular endothelial growth factor receptor FLK1/KDR and PDGFRbeta. Sunitinib 90-97 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 194-197 12748309-9 2003 These results expand the profile of SU11248 as a KIT signaling inhibitor and suggest that SU11248 may have clinical potential in the treatment of SCLC via direct antitumor activity mediated via KIT as well as tumor angiogenesis via vascular endothelial growth factor receptor FLK1/KDR and PDGFRbeta. Sunitinib 90-97 kinase insert domain receptor Homo sapiens 276-280 12748309-9 2003 These results expand the profile of SU11248 as a KIT signaling inhibitor and suggest that SU11248 may have clinical potential in the treatment of SCLC via direct antitumor activity mediated via KIT as well as tumor angiogenesis via vascular endothelial growth factor receptor FLK1/KDR and PDGFRbeta. Sunitinib 90-97 kinase insert domain receptor Homo sapiens 281-284 12748309-9 2003 These results expand the profile of SU11248 as a KIT signaling inhibitor and suggest that SU11248 may have clinical potential in the treatment of SCLC via direct antitumor activity mediated via KIT as well as tumor angiogenesis via vascular endothelial growth factor receptor FLK1/KDR and PDGFRbeta. Sunitinib 90-97 platelet derived growth factor receptor beta Homo sapiens 289-298 14713109-0 2003 SU11248 inhibits tumor growth and CSF-1R-dependent osteolysis in an experimental breast cancer bone metastasis model. Sunitinib 0-7 colony stimulating factor 1 receptor Mus musculus 34-40 12538485-0 2003 In vivo antitumor activity of SU11248, a novel tyrosine kinase inhibitor targeting vascular endothelial growth factor and platelet-derived growth factor receptors: determination of a pharmacokinetic/pharmacodynamic relationship. Sunitinib 30-37 vascular endothelial growth factor A Homo sapiens 83-117 12538485-3 2003 SU11248, a novel small molecule receptor tyrosine kinase inhibitor with direct antitumor as well as antiangiogenic activity via targeting the vascular endothelial growth factor (VEGF), platelet-derived growth factor (PDGF), KIT, and FLT3 receptor tyrosine kinases, was used as the pharmacological agent in these studies. Sunitinib 0-7 vascular endothelial growth factor A Homo sapiens 142-176 12538485-3 2003 SU11248, a novel small molecule receptor tyrosine kinase inhibitor with direct antitumor as well as antiangiogenic activity via targeting the vascular endothelial growth factor (VEGF), platelet-derived growth factor (PDGF), KIT, and FLT3 receptor tyrosine kinases, was used as the pharmacological agent in these studies. Sunitinib 0-7 vascular endothelial growth factor A Homo sapiens 178-182 14713109-1 2003 The aim of the study was to investigate inhibitory effects of the receptor tyrosine kinase (RTK) inhibitor SU11248 against CSF-1R and osteoclast (OC) formation. Sunitinib 107-114 colony stimulating factor 1 receptor Mus musculus 123-129 12538485-3 2003 SU11248, a novel small molecule receptor tyrosine kinase inhibitor with direct antitumor as well as antiangiogenic activity via targeting the vascular endothelial growth factor (VEGF), platelet-derived growth factor (PDGF), KIT, and FLT3 receptor tyrosine kinases, was used as the pharmacological agent in these studies. Sunitinib 0-7 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 224-227 12538485-3 2003 SU11248, a novel small molecule receptor tyrosine kinase inhibitor with direct antitumor as well as antiangiogenic activity via targeting the vascular endothelial growth factor (VEGF), platelet-derived growth factor (PDGF), KIT, and FLT3 receptor tyrosine kinases, was used as the pharmacological agent in these studies. Sunitinib 0-7 fms related receptor tyrosine kinase 3 Homo sapiens 233-237 14713109-3 2003 The in vitro effects of SU11248 on CSF-1R phosphorylation, OC formation and function were evaluated. Sunitinib 24-31 colony stimulating factor 1 (macrophage) Mus musculus 35-40 12538485-6 2003 In target modulation studies in vivo, SU11248 selectively inhibited Flk-1/KDR (VEGF receptor 2) and PDGF receptor beta phosphorylation (in a time- and dose-dependent manner) when plasma concentrations of inhibitor reached or exceeded 50-100 ng/ml. Sunitinib 38-45 kinase insert domain receptor Homo sapiens 68-73 14713109-5 2003 Phosphorylation of the receptor for M-CSF (CSF-1R) expressed by NIH3T3 cells was inhibited by SU11248 with an IC50 of 50-100 nM, consistent with CSF-1R belonging to the class III split kinase domain RTK family. Sunitinib 94-101 colony stimulating factor 1 (macrophage) Mus musculus 36-41 12538485-6 2003 In target modulation studies in vivo, SU11248 selectively inhibited Flk-1/KDR (VEGF receptor 2) and PDGF receptor beta phosphorylation (in a time- and dose-dependent manner) when plasma concentrations of inhibitor reached or exceeded 50-100 ng/ml. Sunitinib 38-45 kinase insert domain receptor Homo sapiens 74-77 12538485-6 2003 In target modulation studies in vivo, SU11248 selectively inhibited Flk-1/KDR (VEGF receptor 2) and PDGF receptor beta phosphorylation (in a time- and dose-dependent manner) when plasma concentrations of inhibitor reached or exceeded 50-100 ng/ml. Sunitinib 38-45 vascular endothelial growth factor A Homo sapiens 79-83 14713109-5 2003 Phosphorylation of the receptor for M-CSF (CSF-1R) expressed by NIH3T3 cells was inhibited by SU11248 with an IC50 of 50-100 nM, consistent with CSF-1R belonging to the class III split kinase domain RTK family. Sunitinib 94-101 colony stimulating factor 1 receptor Mus musculus 43-49 14713109-5 2003 Phosphorylation of the receptor for M-CSF (CSF-1R) expressed by NIH3T3 cells was inhibited by SU11248 with an IC50 of 50-100 nM, consistent with CSF-1R belonging to the class III split kinase domain RTK family. Sunitinib 94-101 colony stimulating factor 1 receptor Mus musculus 145-151 14713109-6 2003 The early M-CSF-dependent phase of in vitro murine OC development and function were inhibited by SU11248 at 10-100 nM. Sunitinib 97-104 colony stimulating factor 1 (macrophage) Mus musculus 10-15 14988747-8 2003 Although very preliminary, a phase I trial found tumor regressions that were caused by an oral VEGF receptor tyrosine kinase inhibitor, SU11248. Sunitinib 136-143 vascular endothelial growth factor A Homo sapiens 95-99 33972682-5 2021 TKI sunitinib resistance was rescued by DPP4 inhibition using sitagliptin or specific siRNAs in RCC cells and tumors. Sunitinib 4-13 dipeptidyl peptidase 4 Homo sapiens 40-44 33799686-6 2021 Compared with control cells transduced with scrambled vector (UMRC3-SC cells), PHD3-knockdown cells (UMRC3-PHD3KD cells) showed increased cell invasion, tumor growth, and response to sunitinib. Sunitinib 183-192 egl-9 family hypoxia inducible factor 3 Homo sapiens 79-83 33799686-8 2021 However, following sunitinib treatment, expression of HIF2alpha and phosphorylated EGFR were equivalent in both PHD3 knockdown and control tumors. Sunitinib 19-28 endothelial PAS domain protein 1 Homo sapiens 54-63 33799686-8 2021 However, following sunitinib treatment, expression of HIF2alpha and phosphorylated EGFR were equivalent in both PHD3 knockdown and control tumors. Sunitinib 19-28 epidermal growth factor Homo sapiens 83-87 33762959-9 2021 Compared with the C allele, the ABCB1 rs1128503 T allele was associated with a decreased risk of sunitinib-induced hypertension but worse progression-free survival (PFS) (ES = 0.44, 95% CI = 0.26-0.77, p = 0.004; ES = 1.36, 95% CI = 1.07-1.73, p = 0.011). Sunitinib 97-106 ATP binding cassette subfamily B member 1 Homo sapiens 32-37 33762959-0 2021 Meta-Analysis of ABCG2 and ABCB1 Polymorphisms With Sunitinib-Induced Toxicity and Efficacy in Renal Cell Carcinoma. Sunitinib 52-61 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 17-22 33762959-0 2021 Meta-Analysis of ABCG2 and ABCB1 Polymorphisms With Sunitinib-Induced Toxicity and Efficacy in Renal Cell Carcinoma. Sunitinib 52-61 ATP binding cassette subfamily B member 1 Homo sapiens 27-32 33762959-12 2021 Conclusion: The results indicate that the ABCG2 rs2231142 polymorphism may serve as a predictor of sunitinib-induced thrombocytopenia and HFS in Asians, while the ABCB1 rs1128503 polymorphism may serve as a predictor of sunitinib-induced hypertension, and both the ABCB1 rs1128503 and rs2032582 polymorphisms may serve as predictors of PFS in RCC. Sunitinib 99-108 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 42-47 33762959-1 2021 Background: ABCG2 and ABCB1 are genes related to the pharmacokinetics of sunitinib and have been associated with its toxicity and efficacy. Sunitinib 73-82 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 12-17 33762959-1 2021 Background: ABCG2 and ABCB1 are genes related to the pharmacokinetics of sunitinib and have been associated with its toxicity and efficacy. Sunitinib 73-82 ATP binding cassette subfamily B member 1 Homo sapiens 22-27 33762959-3 2021 This study aimed to evaluate the associations of ABCG2 and ABCB1 polymorphisms with sunitinib-induced toxicity and efficacy in renal cell carcinoma (RCC) by meta-analysis. Sunitinib 84-93 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 49-54 33762959-3 2021 This study aimed to evaluate the associations of ABCG2 and ABCB1 polymorphisms with sunitinib-induced toxicity and efficacy in renal cell carcinoma (RCC) by meta-analysis. Sunitinib 84-93 ATP binding cassette subfamily B member 1 Homo sapiens 59-64 33762959-7 2021 The ABCG2 rs2231142 A allele was associated with an increased risk of sunitinib-induced thrombocytopenia and hand-foot syndrome (HFS) in Asians (ES = 1.65, 95% CI = 1.15-2.36, p = 0.006; ES = 1.52, 95% CI = 1.02-2.27, p = 0.041). Sunitinib 70-79 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 4-9 33762959-12 2021 Conclusion: The results indicate that the ABCG2 rs2231142 polymorphism may serve as a predictor of sunitinib-induced thrombocytopenia and HFS in Asians, while the ABCB1 rs1128503 polymorphism may serve as a predictor of sunitinib-induced hypertension, and both the ABCB1 rs1128503 and rs2032582 polymorphisms may serve as predictors of PFS in RCC. Sunitinib 99-108 ATP binding cassette subfamily B member 1 Homo sapiens 265-270 33762959-12 2021 Conclusion: The results indicate that the ABCG2 rs2231142 polymorphism may serve as a predictor of sunitinib-induced thrombocytopenia and HFS in Asians, while the ABCB1 rs1128503 polymorphism may serve as a predictor of sunitinib-induced hypertension, and both the ABCB1 rs1128503 and rs2032582 polymorphisms may serve as predictors of PFS in RCC. Sunitinib 220-229 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 42-47 33812180-4 2021 As a result of the inhibition that is caused by sunitinib malate agent at the receptor level, vascular endothelial growth factor (VEGF) level increases. Sunitinib 48-57 vascular endothelial growth factor A Homo sapiens 94-128 33812180-4 2021 As a result of the inhibition that is caused by sunitinib malate agent at the receptor level, vascular endothelial growth factor (VEGF) level increases. Sunitinib 48-57 vascular endothelial growth factor A Homo sapiens 130-134 34949839-5 2022 Several other Food and Drug Administration (FDA)-approved inhibitors, such erlotinib and sunitinib, also bound and activated GCN2. Sunitinib 89-98 eukaryotic translation initiation factor 2 alpha kinase 4 Homo sapiens 125-129 24874929-0 2014 Efflux pump ABCB1 single nucleotide polymorphisms and dose reductions in patients with metastatic renal cell carcinoma treated with sunitinib. Sunitinib 132-141 ATP binding cassette subfamily B member 1 Homo sapiens 12-17 24874929-4 2014 We genotyped 11 key SNPs, respectively, in ABCB1, NR1/2, NR1/3 and CYP3A5, involved in sunitinib pharmacokinetics as well as VEGFR1 and VEGFR3, which have been suggested as regulators of sunitinib pharmacodynamics. Sunitinib 87-96 ATP binding cassette subfamily B member 1 Homo sapiens 43-48 24874929-4 2014 We genotyped 11 key SNPs, respectively, in ABCB1, NR1/2, NR1/3 and CYP3A5, involved in sunitinib pharmacokinetics as well as VEGFR1 and VEGFR3, which have been suggested as regulators of sunitinib pharmacodynamics. Sunitinib 87-96 glutamate ionotropic receptor NMDA type subunit 1 Homo sapiens 50-55 24874929-4 2014 We genotyped 11 key SNPs, respectively, in ABCB1, NR1/2, NR1/3 and CYP3A5, involved in sunitinib pharmacokinetics as well as VEGFR1 and VEGFR3, which have been suggested as regulators of sunitinib pharmacodynamics. Sunitinib 87-96 glutamate ionotropic receptor NMDA type subunit 1 Homo sapiens 50-53 24874929-4 2014 We genotyped 11 key SNPs, respectively, in ABCB1, NR1/2, NR1/3 and CYP3A5, involved in sunitinib pharmacokinetics as well as VEGFR1 and VEGFR3, which have been suggested as regulators of sunitinib pharmacodynamics. Sunitinib 87-96 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 67-73 24874929-4 2014 We genotyped 11 key SNPs, respectively, in ABCB1, NR1/2, NR1/3 and CYP3A5, involved in sunitinib pharmacokinetics as well as VEGFR1 and VEGFR3, which have been suggested as regulators of sunitinib pharmacodynamics. Sunitinib 187-196 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 67-73 25639617-0 2015 Refractory angiosarcoma of the breast with VEGFR2 upregulation successfully treated with sunitinib. Sunitinib 89-98 kinase insert domain receptor Homo sapiens 43-49 34826699-2 2022 As interleukin-6 (IL-6) is overexpressed in sunitinib-resistant cells, the purpose of this study was to explore the potential of IL-6 inhibition with tocilizumab, an IL-6 receptor inhibitor, to overcome resistance. Sunitinib 44-53 interleukin 6 Mus musculus 3-16 34826699-2 2022 As interleukin-6 (IL-6) is overexpressed in sunitinib-resistant cells, the purpose of this study was to explore the potential of IL-6 inhibition with tocilizumab, an IL-6 receptor inhibitor, to overcome resistance. Sunitinib 44-53 interleukin 6 Mus musculus 18-22 34826699-2 2022 As interleukin-6 (IL-6) is overexpressed in sunitinib-resistant cells, the purpose of this study was to explore the potential of IL-6 inhibition with tocilizumab, an IL-6 receptor inhibitor, to overcome resistance. Sunitinib 44-53 interleukin 6 Mus musculus 129-133 34826699-7 2022 In vitro, tocilizumab induced significant cell death, and reduced the expression of IL-6, VEGF, and Bcl-2 in sunitinib-resistant cells. Sunitinib 109-118 interleukin 6 Mus musculus 84-88 34826699-7 2022 In vitro, tocilizumab induced significant cell death, and reduced the expression of IL-6, VEGF, and Bcl-2 in sunitinib-resistant cells. Sunitinib 109-118 vascular endothelial growth factor A Mus musculus 90-94 34826699-7 2022 In vitro, tocilizumab induced significant cell death, and reduced the expression of IL-6, VEGF, and Bcl-2 in sunitinib-resistant cells. Sunitinib 109-118 B cell leukemia/lymphoma 2 Mus musculus 100-105 34868230-9 2021 Finally, drugs including arsenic trioxide, cisplatin, Jinfukang, and sunitinib were mined for the treatment of the eight hub genes. Sunitinib 69-78 ELAV like RNA binding protein 2 Homo sapiens 121-124 34158245-2 2022 We hypothesized that adding sunitinib, a tyrosine kinase inhibitor with antitumor and antiangiogenic activity, to an anthracycline and taxane regimen would improve pathologic complete response (pCR) rates to a prespecified endpoint of 45% in patients with HER2-negative LABC or IBC. Sunitinib 28-37 erb-b2 receptor tyrosine kinase 2 Homo sapiens 256-260 34158245-3 2022 METHODS: We conducted a multicenter, phase II trial of neoadjuvant sunitinib with paclitaxel (S+T) followed by doxorubicin and cyclophosphamide plus G-CSF for patients with HER2-negative LABC or IBC. Sunitinib 67-76 erb-b2 receptor tyrosine kinase 2 Homo sapiens 173-177 34158245-14 2022 The addition of sunitinib to neoadjuvant chemotherapy may provide promising incremental benefit for patients with ER positive LABC. Sunitinib 16-25 estrogen receptor 1 Homo sapiens 114-116 34626691-0 2021 Circular RNA circSNX6 promotes sunitinib resistance in renal cell carcinoma through the miR-1184/GPCPD1/ lysophosphatidic acid axis. Sunitinib 31-40 microRNA 1184-1 Homo sapiens 88-96 34626691-7 2021 circSNX6 acts as a molecular "sponge" to relieve the suppressive effect of microRNA (miR)-1184 on its target gene, glycerophosphocholine phosphodiesterase 1 (GPCPD1), which increases intracellular lysophosphatidic acid (LPA) levels and, ultimately, promotes sunitinib resistance in RCC cells. Sunitinib 258-267 microRNA 1184-1 Homo sapiens 75-94 34626691-7 2021 circSNX6 acts as a molecular "sponge" to relieve the suppressive effect of microRNA (miR)-1184 on its target gene, glycerophosphocholine phosphodiesterase 1 (GPCPD1), which increases intracellular lysophosphatidic acid (LPA) levels and, ultimately, promotes sunitinib resistance in RCC cells. Sunitinib 258-267 glycerophosphocholine phosphodiesterase 1 Homo sapiens 115-156 34626691-7 2021 circSNX6 acts as a molecular "sponge" to relieve the suppressive effect of microRNA (miR)-1184 on its target gene, glycerophosphocholine phosphodiesterase 1 (GPCPD1), which increases intracellular lysophosphatidic acid (LPA) levels and, ultimately, promotes sunitinib resistance in RCC cells. Sunitinib 258-267 glycerophosphocholine phosphodiesterase 1 Homo sapiens 158-164 34626691-8 2021 Our findings demonstrated that the circSNX6/miR-1184/GPCPD1 axis had a critical role in regulation of intracellular LPA levels and sunitinib resistance in RCC; they also provide a novel prognostic indicator and promising therapeutic targets. Sunitinib 131-140 microRNA 1184-1 Homo sapiens 44-52 34626691-8 2021 Our findings demonstrated that the circSNX6/miR-1184/GPCPD1 axis had a critical role in regulation of intracellular LPA levels and sunitinib resistance in RCC; they also provide a novel prognostic indicator and promising therapeutic targets. Sunitinib 131-140 glycerophosphocholine phosphodiesterase 1 Homo sapiens 53-59 34875133-12 2022 Interestingly, the PDGFRA protein fusions can be targeted by tyrosine kinase inhibitors such as imatinib, sunitinib and sorafenib. Sunitinib 106-115 platelet derived growth factor receptor alpha Homo sapiens 19-25 34838486-3 2022 Vascular Endothelial Growth Factor inhibitors have different mechanisms of action, targeting either the ligand (e.g. bevacizumab, anti-Vascular Endothelial Growth Factor monoclonal antibody; aflibercept, recombinant anti-Vascular Endothelial Growth Factor fusion protein), or its receptors such as tyrosine kinase inhibitors (e.g. sunitinib or sorafenib). Sunitinib 331-340 vascular endothelial growth factor A Homo sapiens 0-34 34461182-0 2022 Sunitinib increases the cancer stem cells and vasculogenic mimicry formation via modulating the lncRNA-ECVSR/ERbeta/Hif2-alpha signaling. Sunitinib 0-9 estrogen receptor 1 Homo sapiens 109-115 34461182-0 2022 Sunitinib increases the cancer stem cells and vasculogenic mimicry formation via modulating the lncRNA-ECVSR/ERbeta/Hif2-alpha signaling. Sunitinib 0-9 endothelial PAS domain protein 1 Homo sapiens 116-126 34461182-4 2022 Mechanism dissection revealed that sunitinib can increase the expression of a long non-coding RNA (lncRNA), lncRNA-ECVSR, thereby enhancing the stability of estrogen receptor beta (ERbeta) mRNA. Sunitinib 35-44 estrogen receptor 1 Homo sapiens 157-179 34461182-4 2022 Mechanism dissection revealed that sunitinib can increase the expression of a long non-coding RNA (lncRNA), lncRNA-ECVSR, thereby enhancing the stability of estrogen receptor beta (ERbeta) mRNA. Sunitinib 35-44 estrogen receptor 1 Homo sapiens 181-187 34461182-6 2022 Notably, sunitinib-increased lncRNA-ECVSR/ERbeta/Hif2-alpha signaling resulted in an increased cancer stem cell (CSC) phenotype, thereby promoting VM formation. Sunitinib 9-18 estrogen receptor 1 Homo sapiens 42-48 34461182-6 2022 Notably, sunitinib-increased lncRNA-ECVSR/ERbeta/Hif2-alpha signaling resulted in an increased cancer stem cell (CSC) phenotype, thereby promoting VM formation. Sunitinib 9-18 endothelial PAS domain protein 1 Homo sapiens 49-59 34461182-7 2022 Furthermore, the sunitinib/lncRNA-ECVSR-increased ERbeta expression can transcriptionally regulate lncRNA-ECVSR expression via a positive-feedback loop. Sunitinib 17-26 estrogen receptor 1 Homo sapiens 50-56 34881630-0 2022 Blocking stanniocalcin 2 reduces sunitinib resistance in clear cell renal cell carcinoma. Sunitinib 33-42 stanniocalcin 2 Homo sapiens 9-24 34881630-3 2022 In this study, we investigated the relationship between high expression of STC2 and sunitinib resistance in cells and the underlying mechanism. Sunitinib 84-93 stanniocalcin 2 Homo sapiens 75-79 34881630-7 2022 The sunitinib resistance could be significantly reduced by STC2 neutralizing antibody but aggravated by the addition of recombinant human STC2 in ccRCC cells. Sunitinib 4-13 stanniocalcin 2 Homo sapiens 59-63 34881630-7 2022 The sunitinib resistance could be significantly reduced by STC2 neutralizing antibody but aggravated by the addition of recombinant human STC2 in ccRCC cells. Sunitinib 4-13 stanniocalcin 2 Homo sapiens 138-142 34881630-8 2022 Sunitinib suppressed STC2 expression and secretion, destroyed lysosomal acidic pH, and accumulated in the cells. Sunitinib 0-9 stanniocalcin 2 Homo sapiens 21-25 34881630-9 2022 However, STC2 neutralizing antibody can reduce the accumulation of sunitinib in cells to improve the inhibitory efficiency of sunitinib on cell proliferation. Sunitinib 67-76 stanniocalcin 2 Homo sapiens 9-13 34881630-9 2022 However, STC2 neutralizing antibody can reduce the accumulation of sunitinib in cells to improve the inhibitory efficiency of sunitinib on cell proliferation. Sunitinib 126-135 stanniocalcin 2 Homo sapiens 9-13 34857034-0 2021 Correction to: PFKFB4 is overexpressed in clear-cell renal cell carcinoma promoting pentose phosphate pathway that mediates Sunitinib resistance. Sunitinib 124-133 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 4 Homo sapiens 15-21 34912710-9 2021 Moreover, drug sensitivity analysis revealed that subgroup C2(SC-A) had the highest sensitivity to immunotherapy CTLA-4 blockade and the vascular endothelial growth factor receptor (VEGFR) inhibitor sunitinib. Sunitinib 199-208 complement C2 Homo sapiens 59-66 34912710-9 2021 Moreover, drug sensitivity analysis revealed that subgroup C2(SC-A) had the highest sensitivity to immunotherapy CTLA-4 blockade and the vascular endothelial growth factor receptor (VEGFR) inhibitor sunitinib. Sunitinib 199-208 kinase insert domain receptor Homo sapiens 137-180 34912710-9 2021 Moreover, drug sensitivity analysis revealed that subgroup C2(SC-A) had the highest sensitivity to immunotherapy CTLA-4 blockade and the vascular endothelial growth factor receptor (VEGFR) inhibitor sunitinib. Sunitinib 199-208 kinase insert domain receptor Homo sapiens 182-187 34610839-9 2021 The extent of nuclear localization of YAP1 was significantly reduced after sunitinib stimulation and tended to return to normal levels after drug washout. Sunitinib 75-84 Yes1 associated transcriptional regulator Homo sapiens 38-42 34775465-8 2021 Sunitinib inhibited MamBo38HER2loss tumor growth in vivo and reduced stemness and IL6 production in vitro. Sunitinib 0-9 interleukin 6 Homo sapiens 82-85 34669974-11 2022 In addition, the estimated sunitinib apparent oral clearance (CL/F) was significantly lower in the severe adverse effects group. Sunitinib 27-36 crooked neck pre-mRNA splicing factor 1 Homo sapiens 62-66 34681859-12 2021 Dasatinib, fostamatinib, sunitinib, and pazopanib interfered in early collagen receptor-induced molecular-signaling compared with cabozantinib and vatalanib. Sunitinib 25-34 integrin subunit alpha 2 Homo sapiens 70-87 34722286-2 2021 In this study, we reported a novel GNAS mutation and 1p/22q co-deletion responding to sunitinib in a patient with multiple recurrent meningiomas. Sunitinib 86-95 GNAS complex locus Homo sapiens 35-39 34763724-2 2021 In recent years, pneumatosis intestinalis was reported in patients undergoing cancer treatment, and some case reports exist that report that pneumatosis intestinalis occurs during administration of vascular endothelial growth factor inhibitors, such as bevacizumab and sunitinib. Sunitinib 269-278 vascular endothelial growth factor A Homo sapiens 198-232 34634226-0 2022 Expression of Concern: Depletion of lncRNA MALAT1 inhibited sunitinib resistance through regulating miR-362-3p-mediated G3BP1 in renal cell carcinoma. Sunitinib 60-69 metastasis associated lung adenocarcinoma transcript 1 Homo sapiens 43-49 34634226-0 2022 Expression of Concern: Depletion of lncRNA MALAT1 inhibited sunitinib resistance through regulating miR-362-3p-mediated G3BP1 in renal cell carcinoma. Sunitinib 60-69 G3BP stress granule assembly factor 1 Homo sapiens 120-125 34413312-5 2021 A saFRET biosensor-based high-throughput drug screening (saFRET-HTDS) assay further enables the identification of an FDA-approved cancer drug, Sunitinib, that can be repurposed to inhibit ZAP70 activity and autoimmune-disease-related T-cell activation. Sunitinib 143-152 zeta chain of T cell receptor associated protein kinase 70 Homo sapiens 188-193 34402193-6 2021 RESULTS: We refined immature dendritic cells and central memory CD4 T cells which showed associations with sunitinib and ccRCC. Sunitinib 107-116 CD4 molecule Homo sapiens 64-67 34402193-7 2021 Following, five hub genes (CRYBB1, RIMBP3C, CEACAM4, HAMP, and LYL1) were identified for their strong relationships with sunitinib and immune infiltration in ccRCC. Sunitinib 121-130 crystallin beta B1 Homo sapiens 27-33 34402193-7 2021 Following, five hub genes (CRYBB1, RIMBP3C, CEACAM4, HAMP, and LYL1) were identified for their strong relationships with sunitinib and immune infiltration in ccRCC. Sunitinib 121-130 RIMS binding protein 3C Homo sapiens 35-42 34402193-7 2021 Following, five hub genes (CRYBB1, RIMBP3C, CEACAM4, HAMP, and LYL1) were identified for their strong relationships with sunitinib and immune infiltration in ccRCC. Sunitinib 121-130 CEA cell adhesion molecule 4 Homo sapiens 44-51 34402193-7 2021 Following, five hub genes (CRYBB1, RIMBP3C, CEACAM4, HAMP, and LYL1) were identified for their strong relationships with sunitinib and immune infiltration in ccRCC. Sunitinib 121-130 hepcidin antimicrobial peptide Homo sapiens 53-57 34402193-7 2021 Following, five hub genes (CRYBB1, RIMBP3C, CEACAM4, HAMP, and LYL1) were identified for their strong relationships with sunitinib and immune infiltration in ccRCC. Sunitinib 121-130 LYL1 basic helix-loop-helix family member Homo sapiens 63-67 34402193-8 2021 Further validations in external data refined CRYBB1, CEACAM4, and HAMP which play a vital role in sunitinib resistance, immune infiltrations in ccRCC, and the development and progression of ccRCC. Sunitinib 98-107 crystallin beta B1 Homo sapiens 45-51 34402193-8 2021 Further validations in external data refined CRYBB1, CEACAM4, and HAMP which play a vital role in sunitinib resistance, immune infiltrations in ccRCC, and the development and progression of ccRCC. Sunitinib 98-107 CEA cell adhesion molecule 4 Homo sapiens 53-60 34765076-5 2021 We found that S6 activation was more common in poor risk NC-RCC tumors and S6/Akt activation was associated with worse PFS and OS outcomes with both everolimus and sunitinib, while c-kit was commonly expressed in chromophobe tumors and associated with improved outcomes with both agents. Sunitinib 164-173 AKT serine/threonine kinase 1 Homo sapiens 78-81 34549873-14 2021 Within the translational endpoints, pre-surgical Sunitinib significantly increased necrosis, and reduced CD31, Ki67, circulating VEGF-C, and Ktrans (measured using DCE-MRI) (all p<0.05). Sunitinib 49-58 platelet and endothelial cell adhesion molecule 1 Homo sapiens 105-109 34549873-14 2021 Within the translational endpoints, pre-surgical Sunitinib significantly increased necrosis, and reduced CD31, Ki67, circulating VEGF-C, and Ktrans (measured using DCE-MRI) (all p<0.05). Sunitinib 49-58 vascular endothelial growth factor C Homo sapiens 129-135 34545713-8 2021 RESULTS: Four kinase inhibitors (lapatinib, PD169316, sunitinib, gefitinib) significantly increased ALA-PpIX fluorescence and PDT response in TNBC cells with ABCG2 activity, but not in MCF10A nontumor breast epithelial cell line without ABCG2 activity. Sunitinib 54-63 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 158-163 34236401-2 2021 KIT inhibitors commonly used to treat GIST (eg, imatinib and sunitinib) are inactive-state (type II) inhibitors. Sunitinib 61-70 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 0-3 34216806-5 2021 RESULTS: Our study showed that treatment with sunitinib and photoirradiation at 8 mW/cm2 for 30 min resulted in the production intracellular reactive oxygen species (ROS), which is indicated by the increase in mRNA expression levels of PAI-1, NF-kappabeta, and Caspase-3. Sunitinib 46-55 serpin family E member 1 Homo sapiens 236-241 34216806-5 2021 RESULTS: Our study showed that treatment with sunitinib and photoirradiation at 8 mW/cm2 for 30 min resulted in the production intracellular reactive oxygen species (ROS), which is indicated by the increase in mRNA expression levels of PAI-1, NF-kappabeta, and Caspase-3. Sunitinib 46-55 nuclear factor kappa B subunit 1 Homo sapiens 243-255 34216806-5 2021 RESULTS: Our study showed that treatment with sunitinib and photoirradiation at 8 mW/cm2 for 30 min resulted in the production intracellular reactive oxygen species (ROS), which is indicated by the increase in mRNA expression levels of PAI-1, NF-kappabeta, and Caspase-3. Sunitinib 46-55 caspase 3 Homo sapiens 261-270 34503211-7 2021 The immunotherapy target PD-L1 was consistently up-regulated in resistant cells, and we demonstrated that the silencing of this gene resulted in an increase in sensitivity to sunitinib treatment only in 786-O-resistant cells, suggesting that some ccRCC patients might benefit from combination therapy with PD-L1 checkpoint inhibitors. Sunitinib 175-184 CD274 molecule Homo sapiens 25-30 34593435-0 2021 CD44 Is Involved in Sunitinib Resistance and Poor Progression-free Survival After Sunitinib Treatment of Renal Cell Carcinoma. Sunitinib 20-29 CD44 molecule (Indian blood group) Homo sapiens 0-4 34593435-0 2021 CD44 Is Involved in Sunitinib Resistance and Poor Progression-free Survival After Sunitinib Treatment of Renal Cell Carcinoma. Sunitinib 82-91 CD44 molecule (Indian blood group) Homo sapiens 0-4 34593435-2 2021 This study aimed to examine the role of CD44 in sunitinib resistance and as a predictive marker in mRCC. Sunitinib 48-57 CD44 molecule (Indian blood group) Homo sapiens 40-44 34593435-3 2021 MATERIALS AND METHODS: We analyzed the effect of CD44 knockdown on sunitinib resistance in RCC cell lines using WST-1 assays. Sunitinib 67-76 CD44 molecule (Indian blood group) Homo sapiens 49-53 34593435-4 2021 CD44 expression in mRCC patients treated with first-line sunitinib was determined by immunohistochemistry. Sunitinib 57-66 CD44 molecule (Indian blood group) Homo sapiens 0-4 34593435-6 2021 RESULTS: CD44 knockdown increased sensitivity to sunitinib. Sunitinib 49-58 CD44 molecule (Indian blood group) Homo sapiens 9-13 34593435-8 2021 CD44-positive cases were associated with poor progression-free survival (PFS) after first-line sunitinib treatment. Sunitinib 95-104 CD44 molecule (Indian blood group) Homo sapiens 0-4 34593435-9 2021 In the JAVELIN 101 study, high CD44 expression was significantly associated with poor PFS after sunitinib but not after avelumab + axitinib therapy. Sunitinib 96-105 CD44 molecule (Indian blood group) Homo sapiens 31-35 34593435-10 2021 CONCLUSION: CD44 is involved in sunitinib resistance and may be a promising marker for sunitinib treatment in mRCC. Sunitinib 32-41 CD44 molecule (Indian blood group) Homo sapiens 12-16 34593435-10 2021 CONCLUSION: CD44 is involved in sunitinib resistance and may be a promising marker for sunitinib treatment in mRCC. Sunitinib 87-96 CD44 molecule (Indian blood group) Homo sapiens 12-16 34402193-8 2021 Further validations in external data refined CRYBB1, CEACAM4, and HAMP which play a vital role in sunitinib resistance, immune infiltrations in ccRCC, and the development and progression of ccRCC. Sunitinib 98-107 hepcidin antimicrobial peptide Homo sapiens 66-70 34593007-0 2021 PFKFB4 is overexpressed in clear-cell renal cell carcinoma promoting pentose phosphate pathway that mediates Sunitinib resistance. Sunitinib 109-118 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 4 Mus musculus 0-6 34593007-11 2021 PFKFB4-knockdown overcame resistance to Sunitinib in vitro and in vivo both in xenograft and tail-vein injection murine models. Sunitinib 40-49 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 4 Mus musculus 0-6 34593007-13 2021 Targeting PFKFB4 held promise to combat resistance to Sunitinib. Sunitinib 54-63 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 4 Mus musculus 10-16 34568318-5 2021 We found that the MEK inhibitor trametinib and the multi-target tyrosine kinase inhibitor sunitinib exquisitely increased apoptotic priming in an NRAS-mutant and in a KMT2A-rearranged cell line presenting a high expression of FLT3, respectively. Sunitinib 90-99 NRAS proto-oncogene, GTPase Homo sapiens 146-150 34568318-5 2021 We found that the MEK inhibitor trametinib and the multi-target tyrosine kinase inhibitor sunitinib exquisitely increased apoptotic priming in an NRAS-mutant and in a KMT2A-rearranged cell line presenting a high expression of FLT3, respectively. Sunitinib 90-99 lysine methyltransferase 2A Homo sapiens 167-172 34568318-5 2021 We found that the MEK inhibitor trametinib and the multi-target tyrosine kinase inhibitor sunitinib exquisitely increased apoptotic priming in an NRAS-mutant and in a KMT2A-rearranged cell line presenting a high expression of FLT3, respectively. Sunitinib 90-99 fms related receptor tyrosine kinase 3 Homo sapiens 226-230 34568318-10 2021 Similar observations were made in BCP-ALL KMT2A-rearranged PDX cells in response to sunitinib, showing an analogous DBP profile to the SEM cell line. Sunitinib 84-93 lysine methyltransferase 2A Homo sapiens 42-47 34572331-0 2021 Transgelin Contributes to a Poor Response of Metastatic Renal Cell Carcinoma to Sunitinib Treatment. Sunitinib 80-89 transgelin Homo sapiens 0-10 34572331-9 2021 Our data indicate that transgelin is an essential protein supporting RCC cell proliferation, which could contribute to intrinsic sunitinib resistance. Sunitinib 129-138 transgelin Homo sapiens 23-33 34319001-0 2021 RRM2 Regulates Sensitivity to Sunitinib and PD-1 Blockade in Renal Cancer by Stabilizing ANXA1 and Activating the AKT Pathway. Sunitinib 30-39 ribonucleotide reductase regulatory subunit M2 Homo sapiens 0-4 34319001-8 2021 In this study, the authors have demonstrated that RRM2 is upregulated in sunitinib-resistant RCC cells and patient tissues. Sunitinib 73-82 ribonucleotide reductase regulatory subunit M2 Homo sapiens 50-54 34319001-9 2021 They also find that RRM2 stabilizes ANXA1 and activates the AKT pathway independent of its ribonucleotide reductase activity, promoting sunitinib resistance in RCC. Sunitinib 136-145 ribonucleotide reductase regulatory subunit M2 Homo sapiens 20-24 34475048-0 2021 P53 Is Involved in Sunitinib Resistance and Poor Progression-free Survival After Sunitinib Treatment of Renal Cell Carcinoma. Sunitinib 19-28 tumor protein p53 Homo sapiens 0-3 34475048-0 2021 P53 Is Involved in Sunitinib Resistance and Poor Progression-free Survival After Sunitinib Treatment of Renal Cell Carcinoma. Sunitinib 81-90 tumor protein p53 Homo sapiens 0-3 34475048-3 2021 MATERIALS AND METHODS: We analysed the effects of p53 knockout on sunitinib resistance. Sunitinib 66-75 tumor protein p53 Homo sapiens 50-53 34475048-4 2021 p53 expression in 53 mRCC patients receiving first-line sunitinib was determined immunohistochemically. Sunitinib 56-65 tumor protein p53 Homo sapiens 0-3 34475048-6 2021 RESULTS: WST-1 assays showed that p53 knockout decreased sensitivity to sunitinib. Sunitinib 72-81 tumor protein p53 Homo sapiens 34-37 34475048-9 2021 Kaplan-Meier analysis showed that p53-positive cases tended to be associated with poor progression-free survival (PFS) after first-line sunitinib treatment. Sunitinib 136-145 tumor protein p53 Homo sapiens 34-37 34475048-10 2021 In the JAVELIN 101 study, TP53 mutation was significantly associated with poor PFS after sunitinib treatment. Sunitinib 89-98 tumor protein p53 Homo sapiens 26-30 34475048-11 2021 CONCLUSION: p53 may be involved in sunitinib resistance and represent a valuable marker for sunitinib treatment in mRCC. Sunitinib 35-44 tumor protein p53 Homo sapiens 12-15 34475048-11 2021 CONCLUSION: p53 may be involved in sunitinib resistance and represent a valuable marker for sunitinib treatment in mRCC. Sunitinib 92-101 tumor protein p53 Homo sapiens 12-15 34243074-6 2021 Kinase inhibitory properties support their multi-targeted inhibitory activities against VEGFR-2 and c-kit in similar behavior to that of Sunitinib. Sunitinib 137-146 kinase insert domain receptor Homo sapiens 88-95 34243074-6 2021 Kinase inhibitory properties support their multi-targeted inhibitory activities against VEGFR-2 and c-kit in similar behavior to that of Sunitinib. Sunitinib 137-146 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 100-105 34432562-0 2021 Autophagic degradation of CCN2 (cellular communication network factor 2) causes cardiotoxicity of sunitinib. Sunitinib 98-107 cellular communication network factor 2 Mus musculus 26-30 34432562-0 2021 Autophagic degradation of CCN2 (cellular communication network factor 2) causes cardiotoxicity of sunitinib. Sunitinib 98-107 cellular communication network factor 2 Mus musculus 32-71 34432562-3 2021 Here, we found that sunitinib-induced autophagy leads to apoptosis of cardiomyocyte and cardiac dysfunction as the cardiomyocyte-specific Atg7-/+ heterozygous mice are resistant to sunitinib. Sunitinib 20-29 autophagy related 7 Mus musculus 138-142 34432562-5 2021 Remarkably, deletion of Hmgb1 (high mobility group box 1) inhibited sunitinib-induced cardiomyocyte autophagy and apoptosis, and the HMGB1-specific inhibitor glycyrrhizic acid (GA) significantly mitigated sunitinib-induced autophagy, cardiomyocyte death and cardiotoxicity. Sunitinib 68-77 high mobility group box 1 Mus musculus 24-29 34432562-6 2021 Our study reveals a novel target protein of autophagic degradation in the regulation of cardiomyocyte death and highlights the pharmacological inhibitor of HMGB1 as an attractive approach for improving the safety of sunitinib-based cancer therapy. Sunitinib 216-225 high mobility group box 1 Mus musculus 156-161 34367125-10 2021 We found that Sunitinib significantly decreased the endometriotic lesion size and weight after 1 and 3 weeks, and decreased p-STAT3 activation in MDSCs after 1 week of treatment. Sunitinib 14-23 signal transducer and activator of transcription 3 Homo sapiens 126-131 34363384-0 2021 Lefty A is involved in sunitinib resistance of renal cell carcinoma cells via regulation of IL-8. Sunitinib 23-32 left-right determination factor 2 Homo sapiens 0-7 34363384-8 2021 Knockdown of IL-8 abolished Lefty-regulated sunitinib sensitivity of RCC cells. Sunitinib 44-53 C-X-C motif chemokine ligand 8 Homo sapiens 13-17 34363384-10 2021 Collectively, our present revealed that Lefty A can regulate sunitinib sensitivity of RCC cells of via NF-kappaB/IL-8 signals. Sunitinib 61-70 left-right determination factor 2 Homo sapiens 40-47 34363384-10 2021 Collectively, our present revealed that Lefty A can regulate sunitinib sensitivity of RCC cells of via NF-kappaB/IL-8 signals. Sunitinib 61-70 nuclear factor kappa B subunit 1 Homo sapiens 103-112 34380680-5 2021 Cessation of sunitinib led to reduction in anti-PLA2R antibody IgG titres while resumption, due to concern for cancer progression, led to worsening symptoms. Sunitinib 13-22 phospholipase A2 receptor 1 Homo sapiens 48-53 34367125-11 2021 In the first week, Sunitinib specifically increased the G-MDSC population in peritoneal fluid but the isolated CD11b+Ly6G+Ly6Clo MDSCs after Sunitinib treatment were presented as mature polynuclear MDSCs, while the control group had immature mononuclear MDSCs. Sunitinib 141-150 integrin subunit alpha M Homo sapiens 111-116 34298757-0 2021 New Target for Precision Medicine Treatment of Giant-Cell Tumor of Bone: Sunitinib Is Effective in the Treatment of Neoplastic Stromal Cells with Activated PDGFRbeta Signaling. Sunitinib 73-82 platelet derived growth factor receptor beta Homo sapiens 156-165 34116693-0 2021 Retraction Note to: Effects of HDM2 antagonism on sunitinib resistance, p53 activation, SDF-1 induction, and tumor infiltration by CD11b+/Gr-1+ myeloid derived suppressor cells. Sunitinib 50-59 MDM2 proto-oncogene Homo sapiens 31-35 34306023-0 2021 Identification of MX2 as a Novel Prognostic Biomarker for Sunitinib Resistance in Clear Cell Renal Cell Carcinoma. Sunitinib 58-67 MX dynamin like GTPase 2 Homo sapiens 18-21 34306023-1 2021 Background: Antiangiogenic agents that specifically target vascular endothelial growth factor receptor (VEGFR), such as sunitinib, have been utilized as the standard therapy for metastatic clear cell renal cell carcinoma (ccRCC) patients. Sunitinib 120-129 kinase insert domain receptor Homo sapiens 59-102 34306023-1 2021 Background: Antiangiogenic agents that specifically target vascular endothelial growth factor receptor (VEGFR), such as sunitinib, have been utilized as the standard therapy for metastatic clear cell renal cell carcinoma (ccRCC) patients. Sunitinib 120-129 kinase insert domain receptor Homo sapiens 104-109 34306023-10 2021 Sunitinib-resistant cell lines were constructed, and loss-of-function experiments were conducted by knocking down MX2. Sunitinib 0-9 MX dynamin like GTPase 2 Homo sapiens 114-117 34306023-12 2021 Results: A total of 760 DEGs were derived from two datasets in GEO database, and five hub genes were identified, among which high-level MX2 exhibited a pronounced correlation with poor overall survival (OS) in sunitinib-resistant ccRCC patients. Sunitinib 210-219 MX dynamin like GTPase 2 Homo sapiens 136-139 34306023-14 2021 qRT-PCR and Western blotting revealed that MX2 was distinctly upregulated in sunitinib-resistant RCC cell lines. Sunitinib 77-86 MX dynamin like GTPase 2 Homo sapiens 43-46 34306023-15 2021 Colony formation assay and Cell Counting Kit-8 (CCK8) assay showed that MX2 strongly promoted resistant capability to sunitinib of ccRCC cells. Sunitinib 118-127 MX dynamin like GTPase 2 Homo sapiens 72-75 34306023-16 2021 Conclusion: MX2 is a potent indicator for sunitinib resistance and a therapeutic target in ccRCC patients. Sunitinib 42-51 MX dynamin like GTPase 2 Homo sapiens 12-15 34220265-9 2021 However, hypermethylated seed genes HMGA1 and PSAT1 showcased a good interaction affinity with drugs cisplatin, cyclosporin, bisphenol A, progesterone, and sunitinib, and are crucial in the proliferation of ovarian cancer. Sunitinib 156-165 high mobility group AT-hook 1 Homo sapiens 36-41 34220265-9 2021 However, hypermethylated seed genes HMGA1 and PSAT1 showcased a good interaction affinity with drugs cisplatin, cyclosporin, bisphenol A, progesterone, and sunitinib, and are crucial in the proliferation of ovarian cancer. Sunitinib 156-165 phosphoserine aminotransferase 1 Homo sapiens 46-51 34220265-10 2021 Conclusion: Our study reveals that HMGA1 and PSAT1 can be deployed for initial screening of ovarian cancer and drugs cisplatin, bisphenol A, cyclosporin, progesterone, and sunitinib are effective in curbing the epigenetic alteration. Sunitinib 172-181 high mobility group AT-hook 1 Homo sapiens 35-40 34220265-10 2021 Conclusion: Our study reveals that HMGA1 and PSAT1 can be deployed for initial screening of ovarian cancer and drugs cisplatin, bisphenol A, cyclosporin, progesterone, and sunitinib are effective in curbing the epigenetic alteration. Sunitinib 172-181 phosphoserine aminotransferase 1 Homo sapiens 45-50 34221997-0 2021 Sunitinib Treatment for Advanced Paraganglioma: Case Report of a Novel SDHD Gene Mutation Variant and Systematic Review of the Literature. Sunitinib 0-9 succinate dehydrogenase complex subunit D Homo sapiens 71-75 34221997-4 2021 We describe the case of a man with Familial Paraganglioma Syndrome type 1 (FPGL) related to a novel mutation in SDHD gene treated with sunitinib. Sunitinib 135-144 succinate dehydrogenase complex subunit D Homo sapiens 112-116 34178667-0 2021 Polydatin Reduces Cardiotoxicity and Enhances the Anticancer Effects of Sunitinib by Decreasing Pro-Oxidative Stress, Pro-Inflammatory Cytokines, and NLRP3 Inflammasome Expression. Sunitinib 72-81 NLR family pyrin domain containing 3 Homo sapiens 150-155 34853223-5 2021 We assessed 24 cases who received VEGFR-TKI monotherapy (sunitinib, sorafenib, pazopanib, axitinib) with left ventricular ejection fraction (LVEF) above 50% during the therapy at the Osaka University Hospital from January 2008 to June 2019. Sunitinib 57-66 kinase insert domain receptor Homo sapiens 34-39 34200459-12 2021 In conclusion, high plasma HGF, CXCL11, CXCL10 and IL-6 levels are associated with worse outcome in mRCC patients treated with sunitinib or pazopanib. Sunitinib 127-136 hepatocyte growth factor Homo sapiens 27-30 34200459-12 2021 In conclusion, high plasma HGF, CXCL11, CXCL10 and IL-6 levels are associated with worse outcome in mRCC patients treated with sunitinib or pazopanib. Sunitinib 127-136 C-X-C motif chemokine ligand 11 Homo sapiens 32-38 34200459-12 2021 In conclusion, high plasma HGF, CXCL11, CXCL10 and IL-6 levels are associated with worse outcome in mRCC patients treated with sunitinib or pazopanib. Sunitinib 127-136 C-X-C motif chemokine ligand 10 Homo sapiens 40-46 34200459-12 2021 In conclusion, high plasma HGF, CXCL11, CXCL10 and IL-6 levels are associated with worse outcome in mRCC patients treated with sunitinib or pazopanib. Sunitinib 127-136 interleukin 6 Homo sapiens 51-55 34190510-4 2021 MATERIALS AND METHODS: We analyzed the expression levels of tumor miR-99b, -144, -155, -210, -222, -302a, -377 in 93 RCC patients who received pazopanib or sunitinib in neoadjuvant regimen using RT-PCR. Sunitinib 156-165 microRNA 99b Homo sapiens 66-104 34190510-8 2021 Similarly, the characteristic expression profile of miR-210, -222, -302a and -377 was suggested for sunitinib responders. Sunitinib 100-109 microRNA 210 Homo sapiens 52-59 34121685-2 2021 This study evaluates ezrin expression in sunitinib-treated metastatic clear cell renal cell carcinoma (ccRCC) patients and elucidates its role as a possible marker for survival. Sunitinib 41-50 ezrin Homo sapiens 21-26 34121685-3 2021 Materials and Methods: The expression of ezrin was measured by immunohistochemistry in 80 patients with ccRCC treated by first-line sunitinib between January 2007 and June 2012. Sunitinib 132-141 ezrin Homo sapiens 41-46 34473615-6 2021 RESULTS: Imatinib exhibited the strongest inhibitory effect towards CYP3A, while the inhibitory potential of gefitinib and sunitinib were comparable to each other but weaker than imatinib. Sunitinib 123-132 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 68-73 34473615-12 2021 Therefore, caution should be taken when CYP3A-metabolizing drugs are co-administrated with imatinib, sunitinib, or gefitinib. Sunitinib 101-110 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 40-45 34285770-10 2021 While Sunitinib has already been known to inhibit LRRK2, Crizotinib is a novel LRRK2 binder. Sunitinib 6-15 leucine rich repeat kinase 2 Homo sapiens 50-55 35500680-2 2022 Systemic chemotherapy such as cisplatin and multi-targeted receptor tyrosine kinase inhibitors, including sunitinib, has marginal activity and frequent toxicity. Sunitinib 106-115 ret proto-oncogene Homo sapiens 59-83 35500680-9 2022 Low concentrations of 4 mug/ml cisplatin and 2.8 mug/ml sunitinib showed significant Bcl-2 down-regulation and Bax up-regulation. Sunitinib 56-65 BCL2 apoptosis regulator Homo sapiens 85-90 35443205-0 2022 Pharmacological PDGFRbeta inhibitors imatinib and sunitinib cause human brain pericyte death in vitro. Sunitinib 50-59 platelet derived growth factor receptor beta Homo sapiens 16-25 35500680-9 2022 Low concentrations of 4 mug/ml cisplatin and 2.8 mug/ml sunitinib showed significant Bcl-2 down-regulation and Bax up-regulation. Sunitinib 56-65 BCL2 associated X, apoptosis regulator Homo sapiens 111-114 35358627-1 2022 The efficacy/safety of combining palbociclib (a CDK4/6 inhibitor) and sunitinib (a multi-targeted receptor tyrosine kinase inhibitor) was evaluated, using patient-derived xenograft (PDX) models. Sunitinib 70-79 ret proto-oncogene Homo sapiens 98-122 35266920-1 2022 ABSTRACT: Sunitinib is associated with cardiotoxicity through inhibition of AMP-protein kinase (AMPK) signalling. Sunitinib 10-19 protein kinase AMP-activated catalytic subunit alpha 2 Homo sapiens 76-94 35266920-1 2022 ABSTRACT: Sunitinib is associated with cardiotoxicity through inhibition of AMP-protein kinase (AMPK) signalling. Sunitinib 10-19 protein kinase AMP-activated catalytic subunit alpha 2 Homo sapiens 96-100 35266920-11 2022 The MTT assay demonstrated an increase in the EC50-value during co-administration of metformin with sunitinib compared to sunitinib mono-therapy in HepG2 and HL-60 cell lines, demonstrating the impact and complexity of metformin co-administration and the possible role of AMPK signalling. Sunitinib 100-109 protein kinase AMP-activated catalytic subunit alpha 2 Homo sapiens 272-276 35443205-7 2022 Imatinib and sunitinib, but not orantinib, inhibited PDGFRbeta phosphorylation in pericytes. Sunitinib 13-22 platelet derived growth factor receptor beta Homo sapiens 53-62 35585158-1 2022 Although KIT-mutant GISTs can be effectively treated with tyrosine kinase inhibitors (TKIs), many patients develop resistance to imatinib mesylate (IM) as well as the FDA-approved later-line agents sunitinib, regorafenib and ripretinib. Sunitinib 198-207 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 9-12 35623140-3 2022 Docking studies in the active site of CDK2, one of the key checkpoints enzymes, revealed that the binding scores of the designed molecules are comparable to the reference enzyme"s inhibitors Sunitinib, Nintedanib, and Semaxanib. Sunitinib 191-200 cyclin dependent kinase 2 Homo sapiens 38-42 35538475-0 2022 N6-methyladenosine-modified TRAF1 promotes sunitinib resistance by regulating apoptosis and angiogenesis in a METTL14-dependent manner in renal cell carcinoma. Sunitinib 43-52 TNF receptor associated factor 1 Homo sapiens 28-33 35562668-0 2022 RNF7 inhibits apoptosis and sunitinib sensitivity and promotes glycolysis in renal cell carcinoma via the SOCS1/JAK/STAT3 feedback loop. Sunitinib 28-37 ring finger protein 7 Homo sapiens 0-4 35562668-7 2022 Moreover, RNF7 overexpression affected the sensitivity of RCC cells to sunitinib. Sunitinib 71-80 ring finger protein 7 Homo sapiens 10-14 35562668-9 2022 CONCLUSION: These results demonstrate that RNF7 inhibited apoptosis, promoted glycolysis, and inhibited sunitinib sensitivity in RCC cells via ubiquitination of SOCS1, thus activating STAT3 signaling. Sunitinib 104-113 ring finger protein 7 Homo sapiens 43-47 35562668-9 2022 CONCLUSION: These results demonstrate that RNF7 inhibited apoptosis, promoted glycolysis, and inhibited sunitinib sensitivity in RCC cells via ubiquitination of SOCS1, thus activating STAT3 signaling. Sunitinib 104-113 suppressor of cytokine signaling 1 Homo sapiens 161-166 35562668-9 2022 CONCLUSION: These results demonstrate that RNF7 inhibited apoptosis, promoted glycolysis, and inhibited sunitinib sensitivity in RCC cells via ubiquitination of SOCS1, thus activating STAT3 signaling. Sunitinib 104-113 signal transducer and activator of transcription 3 Homo sapiens 184-189 35538475-8 2022 Moreover, in vivo adeno-associated virus 9 (AAV9) -mediated transduction of TRAF1 suppressed the sunitinib-induced apoptotic and antiangiogenic effects in the CDX models, whereas knockdown of TRAF1 effectively resensitized the sunitinib-resistant CDXs to sunitinib treatment. Sunitinib 97-106 TNF receptor associated factor 1 Homo sapiens 76-81 35538475-8 2022 Moreover, in vivo adeno-associated virus 9 (AAV9) -mediated transduction of TRAF1 suppressed the sunitinib-induced apoptotic and antiangiogenic effects in the CDX models, whereas knockdown of TRAF1 effectively resensitized the sunitinib-resistant CDXs to sunitinib treatment. Sunitinib 97-106 TNF receptor associated factor 1 Homo sapiens 192-197 35538475-8 2022 Moreover, in vivo adeno-associated virus 9 (AAV9) -mediated transduction of TRAF1 suppressed the sunitinib-induced apoptotic and antiangiogenic effects in the CDX models, whereas knockdown of TRAF1 effectively resensitized the sunitinib-resistant CDXs to sunitinib treatment. Sunitinib 227-236 TNF receptor associated factor 1 Homo sapiens 76-81 35562342-5 2022 Consequently, this axis activates VEGFA/VEGFR2 signaling pathway, resulting in angiogenesis and resistance of tumor cells to sunitinib in ccRCC. Sunitinib 125-134 vascular endothelial growth factor A Homo sapiens 34-39 35562342-5 2022 Consequently, this axis activates VEGFA/VEGFR2 signaling pathway, resulting in angiogenesis and resistance of tumor cells to sunitinib in ccRCC. Sunitinib 125-134 kinase insert domain receptor Homo sapiens 40-46 35538475-8 2022 Moreover, in vivo adeno-associated virus 9 (AAV9) -mediated transduction of TRAF1 suppressed the sunitinib-induced apoptotic and antiangiogenic effects in the CDX models, whereas knockdown of TRAF1 effectively resensitized the sunitinib-resistant CDXs to sunitinib treatment. Sunitinib 227-236 TNF receptor associated factor 1 Homo sapiens 192-197 35538475-0 2022 N6-methyladenosine-modified TRAF1 promotes sunitinib resistance by regulating apoptosis and angiogenesis in a METTL14-dependent manner in renal cell carcinoma. Sunitinib 43-52 methyltransferase 14, N6-adenosine-methyltransferase subunit Homo sapiens 110-117 35538475-8 2022 Moreover, in vivo adeno-associated virus 9 (AAV9) -mediated transduction of TRAF1 suppressed the sunitinib-induced apoptotic and antiangiogenic effects in the CDX models, whereas knockdown of TRAF1 effectively resensitized the sunitinib-resistant CDXs to sunitinib treatment. Sunitinib 255-264 TNF receptor associated factor 1 Homo sapiens 76-81 35538475-8 2022 Moreover, in vivo adeno-associated virus 9 (AAV9) -mediated transduction of TRAF1 suppressed the sunitinib-induced apoptotic and antiangiogenic effects in the CDX models, whereas knockdown of TRAF1 effectively resensitized the sunitinib-resistant CDXs to sunitinib treatment. Sunitinib 255-264 TNF receptor associated factor 1 Homo sapiens 192-197 35538475-5 2022 RESULTS: We identified a tumor necrosis factor receptor-associated factor, TRAF1, that was significantly increased in sunitinib-resistant cells, resistant cell-derived xenograft (CDX-R) models and clinical patients with sunitinib resistance. Sunitinib 118-127 TNF receptor associated factor 1 Homo sapiens 75-80 35538475-9 2022 CONCLUSIONS: Overexpression of TRAF1 promotes sunitinib resistance by modulating apoptotic and angiogenic pathways in a METTL14-dependent manner. Sunitinib 46-55 TNF receptor associated factor 1 Homo sapiens 31-36 35538475-5 2022 RESULTS: We identified a tumor necrosis factor receptor-associated factor, TRAF1, that was significantly increased in sunitinib-resistant cells, resistant cell-derived xenograft (CDX-R) models and clinical patients with sunitinib resistance. Sunitinib 220-229 TNF receptor associated factor 1 Homo sapiens 75-80 35538475-9 2022 CONCLUSIONS: Overexpression of TRAF1 promotes sunitinib resistance by modulating apoptotic and angiogenic pathways in a METTL14-dependent manner. Sunitinib 46-55 methyltransferase 14, N6-adenosine-methyltransferase subunit Homo sapiens 120-127 35538475-6 2022 Silencing TRAF1 increased sunitinib-induced apoptotic and antiangiogenic effects. Sunitinib 26-35 TNF receptor associated factor 1 Homo sapiens 10-15 35538475-10 2022 Targeting TRAF1 and its pathways may be a novel pharmaceutical intervention for sunitinib-treated patients. Sunitinib 80-89 TNF receptor associated factor 1 Homo sapiens 10-15 35538475-7 2022 Mechanistically, the upregulated level of TRAF1 in sunitinib-resistant cells was derived from increased TRAF1 RNA stability, which was caused by an increased level of N6-methyladenosine (m6A) in a METTL14-dependent manner. Sunitinib 51-60 TNF receptor associated factor 1 Homo sapiens 42-47 35538475-7 2022 Mechanistically, the upregulated level of TRAF1 in sunitinib-resistant cells was derived from increased TRAF1 RNA stability, which was caused by an increased level of N6-methyladenosine (m6A) in a METTL14-dependent manner. Sunitinib 51-60 TNF receptor associated factor 1 Homo sapiens 104-109 35538475-7 2022 Mechanistically, the upregulated level of TRAF1 in sunitinib-resistant cells was derived from increased TRAF1 RNA stability, which was caused by an increased level of N6-methyladenosine (m6A) in a METTL14-dependent manner. Sunitinib 51-60 methyltransferase 14, N6-adenosine-methyltransferase subunit Homo sapiens 197-204 35412955-0 2022 Circular RNA Eps15-homology domain-containing protein 2 induce resistance of renal cell carcinoma to sunitinib via microRNA-4731-5p/ABCF2 axis. Sunitinib 101-110 EH domain containing 2 Homo sapiens 13-55 35476809-0 2022 Role of Sam68 in Sunitinib induced renal cell carcinoma apoptosis. Sunitinib 17-26 KH RNA binding domain containing, signal transduction associated 1 Homo sapiens 8-13 35476809-3 2022 This article reveals that Sam68 impacts the sensitivity to sunitinib by mediating the apoptosis of RCC cells. Sunitinib 59-68 KH RNA binding domain containing, signal transduction associated 1 Homo sapiens 26-31 35476809-4 2022 Immunohistochemical staining indicated that the Sam68 expression levels in sunitinib sensitive tumor tissues were markedly higher than those in sunitinib resistant tumor tissues. Sunitinib 75-84 KH RNA binding domain containing, signal transduction associated 1 Homo sapiens 48-53 35476809-5 2022 Sunitinib induced RCC cell apoptosis in a concentration-dependent manner and inhibited the expression of total and phosphorylated Sam68 (p-Sam68). Sunitinib 0-9 KH RNA binding domain containing, signal transduction associated 1 Homo sapiens 130-135 35476809-5 2022 Sunitinib induced RCC cell apoptosis in a concentration-dependent manner and inhibited the expression of total and phosphorylated Sam68 (p-Sam68). Sunitinib 0-9 KH RNA binding domain containing, signal transduction associated 1 Homo sapiens 139-144 35476809-6 2022 Downregulation of Sam68 expression inhibited RCC cell apoptosis induced by sunitinib. Sunitinib 75-84 KH RNA binding domain containing, signal transduction associated 1 Homo sapiens 18-23 35476809-7 2022 While upregulation of Sam68 expression could enhance apoptosis induced by sunitinib. Sunitinib 74-83 KH RNA binding domain containing, signal transduction associated 1 Homo sapiens 22-27 35476809-8 2022 Xenograft models showed that tumors in the Sam68-knockdown group did not shrink as much as those in the control group after treatment with sunitinib for 4 weeks. Sunitinib 139-148 KH RNA binding domain containing, signal transduction associated 1 Homo sapiens 43-48 35476809-9 2022 Together, our results suggest that Sam68 expression is associated with the sensitivity of ccRCC patients to sunitinib. Sunitinib 108-117 KH RNA binding domain containing, signal transduction associated 1 Homo sapiens 35-40 35476809-10 2022 Sam68 may promote cell apoptosis induced by sunitinib, and the Sam68 expression level may be a biomarker for predicting sunitinib sensitivity in ccRCC patients. Sunitinib 44-53 KH RNA binding domain containing, signal transduction associated 1 Homo sapiens 0-5 35476809-10 2022 Sam68 may promote cell apoptosis induced by sunitinib, and the Sam68 expression level may be a biomarker for predicting sunitinib sensitivity in ccRCC patients. Sunitinib 44-53 KH RNA binding domain containing, signal transduction associated 1 Homo sapiens 63-68 35476809-10 2022 Sam68 may promote cell apoptosis induced by sunitinib, and the Sam68 expression level may be a biomarker for predicting sunitinib sensitivity in ccRCC patients. Sunitinib 120-129 KH RNA binding domain containing, signal transduction associated 1 Homo sapiens 63-68 35216667-6 2022 Functionally, re-expressing wild-type FH or PTEN C211S phenocopies an AKT inhibitor in suppressing tumor growth and sensitizing PRCC2 to sunitinib. Sunitinib 137-146 fumarate hydratase Homo sapiens 38-40 35216667-6 2022 Functionally, re-expressing wild-type FH or PTEN C211S phenocopies an AKT inhibitor in suppressing tumor growth and sensitizing PRCC2 to sunitinib. Sunitinib 137-146 phosphatase and tensin homolog Homo sapiens 44-48 35216667-6 2022 Functionally, re-expressing wild-type FH or PTEN C211S phenocopies an AKT inhibitor in suppressing tumor growth and sensitizing PRCC2 to sunitinib. Sunitinib 137-146 AKT serine/threonine kinase 1 Homo sapiens 70-73 35216667-8 2022 Collectively, these findings elucidate a non-metabolic, oncogenic role of fumarate in PRCC2 via direct post-translational modification of PTEN and further reveal potential stratification strategies for patients with FH loss by combinatorial AKTi and sunitinib therapy. Sunitinib 250-259 phosphatase and tensin homolog Homo sapiens 138-142 35629359-0 2022 Endothelial EphrinB2 Regulates Sunitinib Therapy Response in Murine Glioma. Sunitinib 31-40 ephrin B2 Mus musculus 12-20 35290929-7 2022 Interestingly, among the identified lead molecules, compound 17 displayed remarkable VEGFR2 inhibition activity with IC50 value of 19.8 nM compared to Sunitinib (IC50 = 75 nM) and Sorafenib (IC50 = 30 nM). Sunitinib 151-160 kinase insert domain receptor Homo sapiens 85-91 35313079-5 2022 To validate the potential of KDR as a therapeutic target for thyroid cancers, we used the KDR RTK inhibitor sunitinib. Sunitinib 108-117 kinase insert domain receptor Homo sapiens 90-93 35165100-6 2022 pTyr-phosphoproteomic profiles of tumor samples from 4 sunitinib-treated versus 7 control patients revealed 108 significantly up- and 23 downregulated (p<0.05) phosphopeptides for sunitinib-treatment, resulting in an EGFR-centered signaling network. Sunitinib 180-189 epidermal growth factor receptor Homo sapiens 217-221 35387658-0 2022 Correction to: Sunitinib-suppressed miR-452-5p facilitates renal cancer cell invasion and metastasis through modulating SMAD4/SMAD7 signals. Sunitinib 15-24 microRNA 452 Homo sapiens 36-43 35387658-0 2022 Correction to: Sunitinib-suppressed miR-452-5p facilitates renal cancer cell invasion and metastasis through modulating SMAD4/SMAD7 signals. Sunitinib 15-24 SMAD family member 4 Homo sapiens 120-125 35387658-0 2022 Correction to: Sunitinib-suppressed miR-452-5p facilitates renal cancer cell invasion and metastasis through modulating SMAD4/SMAD7 signals. Sunitinib 15-24 SMAD family member 7 Homo sapiens 126-131 35409367-0 2022 Sunitinib and Pterostilbene Combination Treatment Exerts Antitumor Effects in Gastric Cancer via Suppression of PDZD8. Sunitinib 0-9 PDZ domain containing 8 Homo sapiens 112-117 35412955-0 2022 Circular RNA Eps15-homology domain-containing protein 2 induce resistance of renal cell carcinoma to sunitinib via microRNA-4731-5p/ABCF2 axis. Sunitinib 101-110 ATP binding cassette subfamily F member 2 Homo sapiens 132-137 35412955-3 2022 This research assessed the role and mechanism of circular RNA circ Eps15-homology domain-containing protein 2 (EHD2) in the resistance of sunitinib (SU) to RCC. Sunitinib 138-147 EH domain containing 2 Homo sapiens 67-109 35412955-3 2022 This research assessed the role and mechanism of circular RNA circ Eps15-homology domain-containing protein 2 (EHD2) in the resistance of sunitinib (SU) to RCC. Sunitinib 138-147 EH domain containing 2 Homo sapiens 111-115 35412955-3 2022 This research assessed the role and mechanism of circular RNA circ Eps15-homology domain-containing protein 2 (EHD2) in the resistance of sunitinib (SU) to RCC. Sunitinib 149-151 EH domain containing 2 Homo sapiens 67-109 35412955-3 2022 This research assessed the role and mechanism of circular RNA circ Eps15-homology domain-containing protein 2 (EHD2) in the resistance of sunitinib (SU) to RCC. Sunitinib 149-151 EH domain containing 2 Homo sapiens 111-115 35412955-14 2022 In conclusion, circEHD2 enhances RCC resistance to SU via acting as a miR-4731-5p sponge to mediate ABCF2. Sunitinib 51-53 microRNA 4731 Homo sapiens 70-78 35412955-14 2022 In conclusion, circEHD2 enhances RCC resistance to SU via acting as a miR-4731-5p sponge to mediate ABCF2. Sunitinib 51-53 ATP binding cassette subfamily F member 2 Homo sapiens 100-105 35456602-7 2022 As demonstrated by the results of inhibition on multi-receptors by Sunitinib, we confirmed that SN-38/Sunitinib co-loaded micelles to be a treatment modality that could inhibit VEGF and PDGF receptors and enhance the antitumor effect of SN-38 (p < 0.05). Sunitinib 102-111 vascular endothelial growth factor A Homo sapiens 177-181 35370676-4 2022 Molecular dynamics (MD) simulations were performed to delve into the interaction of sunitinib and the identified compound M074-2865 with the kinase domain of HPK1. Sunitinib 84-93 mitogen-activated protein kinase kinase kinase kinase 1 Homo sapiens 158-162 35419359-12 2022 As for therapeutic correlations, SLC1A5 was related to the efficacy of dasatinib, sunitinib, sorafenib, and imatinib but may not predict that of radiotherapy, chemotherapeutic drugs, and immune checkpoints inhibitors (ICIs). Sunitinib 82-91 solute carrier family 1 (neutral amino acid transporter), member 5 Mus musculus 33-39 35386213-3 2022 Methods: In this systematic review and network meta-analysis, we searched PubMed, Embase, and Cochrane Library databases for randomized controlled trials (RCTs) with the time set from database establishment to December 10, 2021, using programmed death factor 1 (PD-1) inhibitors, nivolumab, and sunitinib in the treatment of mRCC. Sunitinib 295-304 programmed cell death 1 Homo sapiens 235-260 35386213-3 2022 Methods: In this systematic review and network meta-analysis, we searched PubMed, Embase, and Cochrane Library databases for randomized controlled trials (RCTs) with the time set from database establishment to December 10, 2021, using programmed death factor 1 (PD-1) inhibitors, nivolumab, and sunitinib in the treatment of mRCC. Sunitinib 295-304 programmed cell death 1 Homo sapiens 262-266 35406411-4 2022 We have also found evidence that sunitinib induces RCC VM formation by up-regulating ERbeta expression. Sunitinib 33-42 estrogen receptor 1 Homo sapiens 85-91 35406411-5 2022 In this study, we further demonstrated that treatment with sunitinib, as well as axitinib, another TKI, could induce ERbeta expression in RCC cell lines. Sunitinib 59-68 estrogen receptor 1 Homo sapiens 117-123 35318312-0 2022 Single-cell RNA-sequencing analysis reveals MYH9 promotes renal cell carcinoma development and sunitinib resistance via AKT signaling pathway. Sunitinib 95-104 myosin heavy chain 9 Homo sapiens 44-48 35318312-0 2022 Single-cell RNA-sequencing analysis reveals MYH9 promotes renal cell carcinoma development and sunitinib resistance via AKT signaling pathway. Sunitinib 95-104 AKT serine/threonine kinase 1 Homo sapiens 120-123 35318312-9 2022 Furthermore, MYH9/AKT axis determined the responses of ccRCC cells to sunitinib treatment and might serve as a biomarker for sunitinib benefits in ccRCC patients. Sunitinib 70-79 myosin heavy chain 9 Homo sapiens 13-17 35318312-9 2022 Furthermore, MYH9/AKT axis determined the responses of ccRCC cells to sunitinib treatment and might serve as a biomarker for sunitinib benefits in ccRCC patients. Sunitinib 70-79 AKT serine/threonine kinase 1 Homo sapiens 18-21 35318312-9 2022 Furthermore, MYH9/AKT axis determined the responses of ccRCC cells to sunitinib treatment and might serve as a biomarker for sunitinib benefits in ccRCC patients. Sunitinib 125-134 myosin heavy chain 9 Homo sapiens 13-17 35318312-9 2022 Furthermore, MYH9/AKT axis determined the responses of ccRCC cells to sunitinib treatment and might serve as a biomarker for sunitinib benefits in ccRCC patients. Sunitinib 125-134 AKT serine/threonine kinase 1 Homo sapiens 18-21 35238167-0 2022 A novel role of TCAIM: suppressing renal carcinoma growth and enhancing its sensitivity to sunitinib. Sunitinib 91-100 T cell activation inhibitor, mitochondrial Homo sapiens 16-21 35241735-0 2022 miR-96-5p targets PTEN to mediate sunitinib resistance in clear cell renal cell carcinoma. Sunitinib 34-43 microRNA 96 Homo sapiens 0-6 35241735-0 2022 miR-96-5p targets PTEN to mediate sunitinib resistance in clear cell renal cell carcinoma. Sunitinib 34-43 phosphatase and tensin homolog Homo sapiens 18-22 35241735-4 2022 Using miRNA profiling, we identified miR-96-5p upregulation in tumors from sunitinib-resistant CCRCC patients. Sunitinib 75-84 microRNA 96 Homo sapiens 37-43 35241735-5 2022 By bioinformatic analysis, PTEN was selected as a potential target of miR-96-5p, which showed low levels in tumors from sunitinib-resistant CCRCC patients. Sunitinib 120-129 phosphatase and tensin homolog Homo sapiens 27-31 35241735-5 2022 By bioinformatic analysis, PTEN was selected as a potential target of miR-96-5p, which showed low levels in tumors from sunitinib-resistant CCRCC patients. Sunitinib 120-129 microRNA 96 Homo sapiens 70-76 35241735-7 2022 Additionally, four-week sunitinib treatment increased miR-96-5p and decreased PTEN only in tumors from a sunitinib-resistant patient-derived xenograft model. Sunitinib 24-33 microRNA 96 Homo sapiens 54-60 35328529-0 2022 LPPR5 Expression in Glioma Affects Growth, Vascular Architecture, and Sunitinib Resistance. Sunitinib 70-79 phospholipid phosphatase related 5 Mus musculus 0-5 35311121-17 2022 In addition, CTD database analysis identified abrine, Benzo (A) Pyrene, bisphenol A, Soman, Sunitinib, Tetrachloroethylene, Valproic Acid as seven targeted therapy drugs that may be effective treatments for seven targeted therapeutics. Sunitinib 92-101 CTD Homo sapiens 13-16 35238167-6 2022 Additionally, TCAIM also enhanced their sensitivity to sunitinib by aggravating apoptosis. Sunitinib 55-64 T cell activation inhibitor, mitochondrial Homo sapiens 14-19 35241735-7 2022 Additionally, four-week sunitinib treatment increased miR-96-5p and decreased PTEN only in tumors from a sunitinib-resistant patient-derived xenograft model. Sunitinib 24-33 phosphatase and tensin homolog Homo sapiens 78-82 35241735-7 2022 Additionally, four-week sunitinib treatment increased miR-96-5p and decreased PTEN only in tumors from a sunitinib-resistant patient-derived xenograft model. Sunitinib 105-114 phosphatase and tensin homolog Homo sapiens 78-82 35281041-0 2022 Elevated SNRPA1, as a Promising Predictor Reflecting Severe Clinical Outcome via Effecting Tumor Immunity for ccRCC, Is Related to Cell Invasion, Metastasis, and Sunitinib Sensitivity. Sunitinib 162-171 small nuclear ribonucleoprotein polypeptide A' Homo sapiens 9-15 35241735-9 2022 Furthermore, we demonstrated that repression of PTEN by miR-96-5p increased cell proliferation and migration in sunitinib-treated cell lines. Sunitinib 112-121 phosphatase and tensin homolog Homo sapiens 48-52 35241735-9 2022 Furthermore, we demonstrated that repression of PTEN by miR-96-5p increased cell proliferation and migration in sunitinib-treated cell lines. Sunitinib 112-121 microRNA 96 Homo sapiens 56-62 35241735-10 2022 These results highlight the direct suppression of PTEN by miR-96-5p and that high miR-96-5p and low PTEN are partially responsible for sunitinib resistance and poor prognosis in CCRCC. Sunitinib 135-144 microRNA 96 Homo sapiens 82-88 35241735-10 2022 These results highlight the direct suppression of PTEN by miR-96-5p and that high miR-96-5p and low PTEN are partially responsible for sunitinib resistance and poor prognosis in CCRCC. Sunitinib 135-144 phosphatase and tensin homolog Homo sapiens 100-104 35091290-12 2022 Furthermore, inhibitory activity of compounds 5a, 5b, 5e1, 5m, 6f and 6j on CDK2 enzyme were tested and revealed that compound 6f, with the N-4-flourobenzyl- 2-oxindole and 3-p-chlorobenzylidene moieties, has a comparable inhibitory activity to the reference drug sunitinib. Sunitinib 264-273 cyclin dependent kinase 2 Homo sapiens 76-80 35281041-6 2022 The knockout expression of SNRPA by sgRNA plasmid inhibited the cell proliferation, migration, and metastasis ability and significantly increased the sensitivity of sunitinib treatment. Sunitinib 165-174 small nuclear ribonucleoprotein polypeptide A Homo sapiens 27-32 35155225-8 2022 Crizotinib and sunitinib induced apoptosis via the mitochondrial pathway, which was characterized by decreasing Bcl2/Bax ratio to dissipate the mitochondrial membrane potential, and increasing apoptotic markers levels. Sunitinib 15-24 BCL2 apoptosis regulator Homo sapiens 112-116 35157045-0 2022 Sunitinib inhibits STAT3 phosphorylation in cardiac muscle and prevents cardiomyopathy in the mdx mouse model of Duchenne muscular dystrophy. Sunitinib 0-9 signal transducer and activator of transcription 3 Mus musculus 19-24 35157045-8 2022 Recently, we have shown that a Food and Drug Administration (FDA)-approved small molecule, sunitinib, a multi-targeted tyrosine kinase inhibitor can mitigate skeletal muscle disease through an increase in myogenic capacity, cell membrane integrity, and improvement of skeletal muscle function via regulation of STAT3-related signaling pathway. Sunitinib 91-100 signal transducer and activator of transcription 3 Homo sapiens 311-316 35157045-11 2022 Our results showed sunitinib treatment reduced STAT3 phosphorylation in the heart muscle of mdx mice, improved cardiac electrical function, increased cardiac output and stroke volume, decreased ventricular hypertrophy, reduced cardiomyocytes membrane damage, fibrotic tissue deposition, and slightly decreased cardiac inflammation. Sunitinib 19-28 signal transducer and activator of transcription 3 Mus musculus 47-52 35123514-13 2022 The gene-drug interaction network stated that CENPF inhibitors, such as Cisplatin, Sunitinib, and Etoposide, might serve as potential drugs for the therapy of ACC. Sunitinib 83-92 centromere protein F Homo sapiens 46-51 34974812-4 2022 In the present study, Sunitinib and Everolimus treatment, respectively, downregulated nc886 expression in a dose-dependent manner in all four renal cancer cell lines. Sunitinib 22-31 vault RNA 2-1 Homo sapiens 86-91 34974812-5 2022 Nc886 overexpression in 786-O cells and ACHN cells significantly reduced the sensitivity of cancer cells to both Sunitinib and Everolimus treatment, respectively, by promoting cell viability and inhibiting cell apoptosis, whereas nc886 silencing increased cancer cell sensitivity. Sunitinib 113-122 vault RNA 2-1 Homo sapiens 0-5 34974812-6 2022 In renal cancer cell line with the highest drug-resistance, 786-O cells, Sunitinib, or Everolimus treatment enhanced the cellular EMT and was further enhanced by nc886 overexpression while attenuated by nc886 silencing. Sunitinib 73-82 IL2 inducible T cell kinase Homo sapiens 130-133 34974812-6 2022 In renal cancer cell line with the highest drug-resistance, 786-O cells, Sunitinib, or Everolimus treatment enhanced the cellular EMT and was further enhanced by nc886 overexpression while attenuated by nc886 silencing. Sunitinib 73-82 vault RNA 2-1 Homo sapiens 162-167 34974812-7 2022 In 786-O cells, nc886 overexpression significantly promoted EMT, ROCK2 phosphorylation, and beta-catenin nucleus translocation under Sunitinib or Everolimus treatment. Sunitinib 133-142 vault RNA 2-1 Homo sapiens 16-21 34974812-7 2022 In 786-O cells, nc886 overexpression significantly promoted EMT, ROCK2 phosphorylation, and beta-catenin nucleus translocation under Sunitinib or Everolimus treatment. Sunitinib 133-142 catenin beta 1 Homo sapiens 92-104 34974812-10 2022 nc886 also promotes renal cancer cell drug-resistance to Sunitinib or Everolimus by promoting EMT through Rock2 phosphorylation-mediated nuclear translocation of beta-catenin. Sunitinib 57-66 vault RNA 2-1 Homo sapiens 0-5 34974812-10 2022 nc886 also promotes renal cancer cell drug-resistance to Sunitinib or Everolimus by promoting EMT through Rock2 phosphorylation-mediated nuclear translocation of beta-catenin. Sunitinib 57-66 IL2 inducible T cell kinase Homo sapiens 94-97 34974812-10 2022 nc886 also promotes renal cancer cell drug-resistance to Sunitinib or Everolimus by promoting EMT through Rock2 phosphorylation-mediated nuclear translocation of beta-catenin. Sunitinib 57-66 catenin beta 1 Homo sapiens 162-174 35209964-8 2022 In the supernatant of FLC-4 cells with sunitinib, the heat shock protein (HSP) 90 was significantly increased. Sunitinib 39-48 heat shock protein 90 alpha family class A member 1 Homo sapiens 54-81 35155225-8 2022 Crizotinib and sunitinib induced apoptosis via the mitochondrial pathway, which was characterized by decreasing Bcl2/Bax ratio to dissipate the mitochondrial membrane potential, and increasing apoptotic markers levels. Sunitinib 15-24 BCL2 associated X, apoptosis regulator Homo sapiens 117-120 35155225-10 2022 Taken together, this study demonstrated that crizotinib- and sunitinib-caused oxidative stress and apoptosis finally impaired hepatic function, which was strongly supported by the histopathological lesions and markedly increased levels of serum alanine aminotransferase, alkaline phosphatase and lactate dehydrogenase. Sunitinib 61-70 glutamic--pyruvic transaminase Homo sapiens 245-269 35066605-0 2022 The multitargeted receptor tyrosine kinase inhibitor sunitinib induces resistance of HER2 positive breast cancer cells to trastuzumab-mediated ADCC. Sunitinib 53-62 erb-b2 receptor tyrosine kinase 2 Homo sapiens 85-89 35153769-4 2021 Our screening revealed that both the bromodomain and extra-terminal domain (BET) inhibitor, I-BET151, and kinase inhibitor, sunitinib, decreased the BCL-2 family protein expression and significantly synergized with venetoclax, enhancing KMT2A-r AML cell line death. Sunitinib 124-133 BCL2 apoptosis regulator Homo sapiens 149-154 35153769-4 2021 Our screening revealed that both the bromodomain and extra-terminal domain (BET) inhibitor, I-BET151, and kinase inhibitor, sunitinib, decreased the BCL-2 family protein expression and significantly synergized with venetoclax, enhancing KMT2A-r AML cell line death. Sunitinib 124-133 lysine methyltransferase 2A Homo sapiens 237-242 35066605-7 2022 Moreover, sunitinib induced downregulation of HER2 on the target cells" surface, changed the morphology and increased adherence of the target cells. Sunitinib 10-19 erb-b2 receptor tyrosine kinase 2 Homo sapiens 46-50 35066605-9 2022 Sunitinib-induced ADCC resistance has been confirmed in a 3D tumor model revealing the prevention of apoptotic cell death (Annexin V staining) in JIMT-1 spheroids co-incubated with NK cells and trastuzumab. Sunitinib 0-9 annexin A5 Homo sapiens 123-132 34983047-8 2022 In contranst, the multi-tyrosine kinase inhibitor sunitinib displays better activity in KIT-exon 9 mutant GISTs compared to others. Sunitinib 50-59 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 88-91 35154527-0 2022 The expression of long non-coding RNA HOTAIR in advanced hepatocellular carcinoma and its prognostic correlation with sunitinib therapy. Sunitinib 118-127 HOX transcript antisense RNA Homo sapiens 38-44 35154527-2 2022 In addition, we also investigated the prognostic correlation between the expression level of lncRNA HOTAIR in tumour tissues and peripheral blood of patients with advanced HCC and sunitinib monotherapy. Sunitinib 180-189 HOX transcript antisense RNA Homo sapiens 100-106 35154527-12 2022 Cox regression analysis indicated that the expression level of lncRNA HOTAIR in tumour tissue and peripheral blood was an independent predictive factor of OS and PFS in patients with advanced HCC treated by sunitinib. Sunitinib 207-216 HOX transcript antisense RNA Homo sapiens 70-76 35154527-14 2022 In addition, the expression level of lncRNA HOTAIR was one of the indicators predicting the effectiveness of sunitinib therapy. Sunitinib 109-118 HOX transcript antisense RNA Homo sapiens 44-50