PMID-sentid Pub_year Sent_text compound_name comp_offset prot_official_name organism prot_offset 34740946-0 2022 Ixazomib induces apoptosis and suppresses proliferation in esophageal squamous cell carcinoma through activation of the c-Myc/NOXA pathway. ixazomib 0-8 MYC proto-oncogene, bHLH transcription factor Homo sapiens 120-125 34740946-0 2022 Ixazomib induces apoptosis and suppresses proliferation in esophageal squamous cell carcinoma through activation of the c-Myc/NOXA pathway. ixazomib 0-8 phorbol-12-myristate-13-acetate-induced protein 1 Homo sapiens 126-130 34740946-6 2022 RT-PCR results showed that the expression of endoplasmic reticulum stress-related gene NOXA and c-Myc significant increase after treatment with ixazomib in ESCC cell. ixazomib 144-152 phorbol-12-myristate-13-acetate-induced protein 1 Homo sapiens 87-91 34740946-6 2022 RT-PCR results showed that the expression of endoplasmic reticulum stress-related gene NOXA and c-Myc significant increase after treatment with ixazomib in ESCC cell. ixazomib 144-152 MYC proto-oncogene, bHLH transcription factor Homo sapiens 96-101 34740946-7 2022 Then we knockdown the NOXA and c-Myc by siRNA, the therapeutic effect of ixazomib markedly decrease, which confirmed that c-Myc/NOXA pathway played a key role in the treatment of ESCC with ixazomib. ixazomib 73-81 phorbol-12-myristate-13-acetate-induced protein 1 Homo sapiens 22-26 34740946-7 2022 Then we knockdown the NOXA and c-Myc by siRNA, the therapeutic effect of ixazomib markedly decrease, which confirmed that c-Myc/NOXA pathway played a key role in the treatment of ESCC with ixazomib. ixazomib 73-81 MYC proto-oncogene, bHLH transcription factor Homo sapiens 31-36 34740946-7 2022 Then we knockdown the NOXA and c-Myc by siRNA, the therapeutic effect of ixazomib markedly decrease, which confirmed that c-Myc/NOXA pathway played a key role in the treatment of ESCC with ixazomib. ixazomib 73-81 MYC proto-oncogene, bHLH transcription factor Homo sapiens 122-127 34740946-7 2022 Then we knockdown the NOXA and c-Myc by siRNA, the therapeutic effect of ixazomib markedly decrease, which confirmed that c-Myc/NOXA pathway played a key role in the treatment of ESCC with ixazomib. ixazomib 73-81 phorbol-12-myristate-13-acetate-induced protein 1 Homo sapiens 128-132 34740946-7 2022 Then we knockdown the NOXA and c-Myc by siRNA, the therapeutic effect of ixazomib markedly decrease, which confirmed that c-Myc/NOXA pathway played a key role in the treatment of ESCC with ixazomib. ixazomib 189-197 phorbol-12-myristate-13-acetate-induced protein 1 Homo sapiens 22-26 34740946-7 2022 Then we knockdown the NOXA and c-Myc by siRNA, the therapeutic effect of ixazomib markedly decrease, which confirmed that c-Myc/NOXA pathway played a key role in the treatment of ESCC with ixazomib. ixazomib 189-197 MYC proto-oncogene, bHLH transcription factor Homo sapiens 31-36 34740946-7 2022 Then we knockdown the NOXA and c-Myc by siRNA, the therapeutic effect of ixazomib markedly decrease, which confirmed that c-Myc/NOXA pathway played a key role in the treatment of ESCC with ixazomib. ixazomib 189-197 MYC proto-oncogene, bHLH transcription factor Homo sapiens 122-127 34740946-7 2022 Then we knockdown the NOXA and c-Myc by siRNA, the therapeutic effect of ixazomib markedly decrease, which confirmed that c-Myc/NOXA pathway played a key role in the treatment of ESCC with ixazomib. ixazomib 189-197 phorbol-12-myristate-13-acetate-induced protein 1 Homo sapiens 128-132 34740946-10 2022 These results suggested that ixazomib is known to suppress proliferation and induce apoptosis in an ESCC cell lines, and this effect was likely mediated by increased activation of the c-Myc/NOXA signaling pathways. ixazomib 29-37 MYC proto-oncogene, bHLH transcription factor Homo sapiens 184-189 34740946-10 2022 These results suggested that ixazomib is known to suppress proliferation and induce apoptosis in an ESCC cell lines, and this effect was likely mediated by increased activation of the c-Myc/NOXA signaling pathways. ixazomib 29-37 phorbol-12-myristate-13-acetate-induced protein 1 Homo sapiens 190-194 34378191-11 2021 FCE showed significant synergistic activity in vitro and in vivo with ixazomib. ixazomib 70-78 ferrochelatase Homo sapiens 0-3 34485305-10 2021 Furthermore, integrated analysis of S100 gene expression and ex vivo drug sensitivity data showed significant negative correlation between expression of S100 family members (S100A8, S100A9, and S100A12) and sensitivity to some drugs used in current MM treatment, including proteasome inhibitors (bortezomib, carfilzomib, and ixazomib) and histone deacetylase inhibitor panobinostat. ixazomib 325-333 S100 calcium binding protein A1 Homo sapiens 36-40 34485305-10 2021 Furthermore, integrated analysis of S100 gene expression and ex vivo drug sensitivity data showed significant negative correlation between expression of S100 family members (S100A8, S100A9, and S100A12) and sensitivity to some drugs used in current MM treatment, including proteasome inhibitors (bortezomib, carfilzomib, and ixazomib) and histone deacetylase inhibitor panobinostat. ixazomib 325-333 S100 calcium binding protein A8 Homo sapiens 174-180 34485305-10 2021 Furthermore, integrated analysis of S100 gene expression and ex vivo drug sensitivity data showed significant negative correlation between expression of S100 family members (S100A8, S100A9, and S100A12) and sensitivity to some drugs used in current MM treatment, including proteasome inhibitors (bortezomib, carfilzomib, and ixazomib) and histone deacetylase inhibitor panobinostat. ixazomib 325-333 S100 calcium binding protein A9 Homo sapiens 182-188 34485305-10 2021 Furthermore, integrated analysis of S100 gene expression and ex vivo drug sensitivity data showed significant negative correlation between expression of S100 family members (S100A8, S100A9, and S100A12) and sensitivity to some drugs used in current MM treatment, including proteasome inhibitors (bortezomib, carfilzomib, and ixazomib) and histone deacetylase inhibitor panobinostat. ixazomib 325-333 S100 calcium binding protein A12 Homo sapiens 194-201 35485606-4 2022 The expression of autophagy proteins (LC3A/B and Beclin-1) and UPS protein (ubiquitin) in MDA-MB-231 and MCF-7 cells following doxorubicin, ixazomib, and/or hydroxychloroquine were determined by western blot. ixazomib 140-148 microtubule associated protein 1 light chain 3 alpha Homo sapiens 38-44 35485606-6 2022 Doxorubicin and ixazomib cotreatment increased Beclin-1 (3.8- and 3.5-fold) and LC3-II expression (13.5- and 1.9-fold) in MDA-MB-231 and MCF-7 cells, respectively. ixazomib 16-24 beclin 1 Homo sapiens 47-55 33647537-0 2021 Ixazomib inhibits myeloma cell proliferation by targeting UBE2K. ixazomib 0-8 ubiquitin conjugating enzyme E2 K Homo sapiens 58-63 33647537-3 2021 The purpose of this study was to investigate the relationship between ixazomib and UBE2K in myeloma cells. ixazomib 70-78 ubiquitin conjugating enzyme E2 K Homo sapiens 83-88 33647537-6 2021 Furthermore, the regulatory effects of ixazomib on UBE2K and its downstream targets were investigated following the overexpression of UBE2K. ixazomib 39-47 ubiquitin conjugating enzyme E2 K Homo sapiens 51-56 33647537-8 2021 Ixazomib significantly increased the expression of HIST1H2BD, MNAT1, NEK3, and TARS2, while decreasing the expression of HSPA1B and UBE2K. ixazomib 0-8 H2B clustered histone 5 Homo sapiens 51-60 33647537-8 2021 Ixazomib significantly increased the expression of HIST1H2BD, MNAT1, NEK3, and TARS2, while decreasing the expression of HSPA1B and UBE2K. ixazomib 0-8 MNAT1 component of CDK activating kinase Homo sapiens 62-67 33647537-8 2021 Ixazomib significantly increased the expression of HIST1H2BD, MNAT1, NEK3, and TARS2, while decreasing the expression of HSPA1B and UBE2K. ixazomib 0-8 NIMA related kinase 3 Homo sapiens 69-73 33647537-8 2021 Ixazomib significantly increased the expression of HIST1H2BD, MNAT1, NEK3, and TARS2, while decreasing the expression of HSPA1B and UBE2K. ixazomib 0-8 threonyl-tRNA synthetase 2, mitochondrial Homo sapiens 79-84 33647537-8 2021 Ixazomib significantly increased the expression of HIST1H2BD, MNAT1, NEK3, and TARS2, while decreasing the expression of HSPA1B and UBE2K. ixazomib 0-8 heat shock protein family A (Hsp70) member 1B Homo sapiens 121-127 33647537-8 2021 Ixazomib significantly increased the expression of HIST1H2BD, MNAT1, NEK3, and TARS2, while decreasing the expression of HSPA1B and UBE2K. ixazomib 0-8 ubiquitin conjugating enzyme E2 K Homo sapiens 132-137 33647537-9 2021 In addition, ixazomib inhibited the proliferation of myeloma cells, blocked cell cycle, induced cell apoptosis, and increased the production of reactive oxygen species by inhibiting UBE2K expression. ixazomib 13-21 ubiquitin conjugating enzyme E2 K Homo sapiens 182-187 33647537-10 2021 Lastly, ixazomib regulates mitosis- and apoptosis-related genes by lowering UBE2K expression. ixazomib 8-16 ubiquitin conjugating enzyme E2 K Homo sapiens 76-81 33647537-11 2021 CONCLUSION: In summary, ixazomib leads to impaired proliferation of myeloma cells by targeting UBE2K. ixazomib 24-32 ubiquitin conjugating enzyme E2 K Homo sapiens 95-100 33310890-7 2021 In subcutaneous tumor models, combination of ixazomib and alectinib prominently induced tumor regression and apoptosis even though the tumors were generated from ALK-rearranged NSCLC cells with non-functional p53. ixazomib 45-53 ALK receptor tyrosine kinase Homo sapiens 162-165 33310890-7 2021 In subcutaneous tumor models, combination of ixazomib and alectinib prominently induced tumor regression and apoptosis even though the tumors were generated from ALK-rearranged NSCLC cells with non-functional p53. ixazomib 45-53 tumor protein p53 Homo sapiens 209-212 33641211-5 2021 METHODS: This is a single-center phase Ib study with a 3+3 design of fulvestrant and the proteasome inhibitor ixazomib (MLN9708) in patients with advanced ER+ breast cancer that was progressing on fulvestrant. ixazomib 110-118 estrogen receptor 1 Homo sapiens 155-157 33670955-0 2021 Oral Proteasomal Inhibitors Ixazomib, Oprozomib, and Delanzomib Upregulate the Function of Organic Anion Transporter 3 (OAT3): Implications in OAT3-Mediated Drug-Drug Interactions. ixazomib 28-36 solute carrier family 22 member 8 Homo sapiens 91-118 33670955-0 2021 Oral Proteasomal Inhibitors Ixazomib, Oprozomib, and Delanzomib Upregulate the Function of Organic Anion Transporter 3 (OAT3): Implications in OAT3-Mediated Drug-Drug Interactions. ixazomib 28-36 solute carrier family 22 member 8 Homo sapiens 120-124 33670955-0 2021 Oral Proteasomal Inhibitors Ixazomib, Oprozomib, and Delanzomib Upregulate the Function of Organic Anion Transporter 3 (OAT3): Implications in OAT3-Mediated Drug-Drug Interactions. ixazomib 28-36 solute carrier family 22 member 8 Homo sapiens 143-147 33670955-4 2021 Therefore, this study investigated the effects of ixazomib, oprozomib, and delanzomib on the expression and transport activity of OAT3 and elucidated the underlying mechanisms. ixazomib 50-58 solute carrier family 22 member 8 Homo sapiens 130-134 33670955-7 2021 Together, our study discovered a novel role of anticancer agents ixazomib, oprozomib, and delanzomib in upregulating OAT3 function, unveiled the proteasome as a promising target for OAT3 regulation, and provided implication of OAT3-mediated drug-drug interactions, which should be warned against during combination therapies with proteasome inhibitor drugs. ixazomib 65-73 solute carrier family 22 member 8 Homo sapiens 117-121 33670955-7 2021 Together, our study discovered a novel role of anticancer agents ixazomib, oprozomib, and delanzomib in upregulating OAT3 function, unveiled the proteasome as a promising target for OAT3 regulation, and provided implication of OAT3-mediated drug-drug interactions, which should be warned against during combination therapies with proteasome inhibitor drugs. ixazomib 65-73 solute carrier family 22 member 8 Homo sapiens 182-186 33670955-7 2021 Together, our study discovered a novel role of anticancer agents ixazomib, oprozomib, and delanzomib in upregulating OAT3 function, unveiled the proteasome as a promising target for OAT3 regulation, and provided implication of OAT3-mediated drug-drug interactions, which should be warned against during combination therapies with proteasome inhibitor drugs. ixazomib 65-73 solute carrier family 22 member 8 Homo sapiens 182-186 32607595-6 2021 Similar results were obtained with next-generation proteasome inhibitors ixazomib and carfilzomib, indicating that induction of HSF2 expression is a general response to proteasome dysfunction. ixazomib 73-81 heat shock transcription factor 2 Homo sapiens 128-132 33296126-0 2021 Ixazomib, an oral proteasome inhibitor, exhibits potential effect in dystrophin-deficient mdx mice. ixazomib 0-8 dystrophin, muscular dystrophy Mus musculus 69-79 33296126-12 2021 There was an increase in the expression of dystrophin and utrophin in the TA and DIA muscles and a reduction in the expression of osteopontin and TGF-beta in the DIA muscle of mdx mice treated with ixazomib. ixazomib 198-206 dystrophin, muscular dystrophy Mus musculus 43-53 33296126-12 2021 There was an increase in the expression of dystrophin and utrophin in the TA and DIA muscles and a reduction in the expression of osteopontin and TGF-beta in the DIA muscle of mdx mice treated with ixazomib. ixazomib 198-206 utrophin Mus musculus 58-66 33296126-12 2021 There was an increase in the expression of dystrophin and utrophin in the TA and DIA muscles and a reduction in the expression of osteopontin and TGF-beta in the DIA muscle of mdx mice treated with ixazomib. ixazomib 198-206 secreted phosphoprotein 1 Mus musculus 130-141 33296126-12 2021 There was an increase in the expression of dystrophin and utrophin in the TA and DIA muscles and a reduction in the expression of osteopontin and TGF-beta in the DIA muscle of mdx mice treated with ixazomib. ixazomib 198-206 transforming growth factor alpha Mus musculus 146-154 33296126-13 2021 Ixazomib was thus shown to increase the expression of dystrophin and utrophin associated with improved pathological and functional changes in the dystrophic muscles of mdx mice. ixazomib 0-8 dystrophin, muscular dystrophy Mus musculus 54-64 33296126-13 2021 Ixazomib was thus shown to increase the expression of dystrophin and utrophin associated with improved pathological and functional changes in the dystrophic muscles of mdx mice. ixazomib 0-8 utrophin Mus musculus 69-77 33406639-0 2021 Activation of Serum/Glucocorticoid Regulated Kinase 1/Nuclear Factor-kappaB Pathway Are Correlated with Low Sensitivity to Bortezomib and Ixazomib in Resistant Multiple Myeloma Cells. ixazomib 138-146 serum/glucocorticoid regulated kinase 1 Homo sapiens 14-53 33406639-7 2021 The SGK1 inhibitor enhances the cytotoxic effects of bortezomib and ixazomib; however, FGFR1 and CCR2 inhibitors do not show such effect in KMS-20 cells. ixazomib 68-76 serum/glucocorticoid regulated kinase 1 Homo sapiens 4-8 33406639-8 2021 Moreover, SGK1 activation induces the phosphorylation of NF-kappaB p65, and an NF-kappaB inhibitor enhances the sensitivity of KMS-20 cells to bortezomib and ixazomib. ixazomib 158-166 serum/glucocorticoid regulated kinase 1 Homo sapiens 10-14 33406639-8 2021 Moreover, SGK1 activation induces the phosphorylation of NF-kappaB p65, and an NF-kappaB inhibitor enhances the sensitivity of KMS-20 cells to bortezomib and ixazomib. ixazomib 158-166 nuclear factor kappa B subunit 1 Homo sapiens 57-66 33406639-10 2021 These results indicate that the activation of the SGK1/NF-kappaB pathway correlates with a low sensitivity to bortezomib and ixazomib, and a combination of bortezomib and ixazomib with an SGK1 or NF-kappaB inhibitor may be involved in the treatment of MM via activation of the SGK1/NF-kappaB pathway. ixazomib 125-133 serum/glucocorticoid regulated kinase 1 Homo sapiens 50-54 33406639-10 2021 These results indicate that the activation of the SGK1/NF-kappaB pathway correlates with a low sensitivity to bortezomib and ixazomib, and a combination of bortezomib and ixazomib with an SGK1 or NF-kappaB inhibitor may be involved in the treatment of MM via activation of the SGK1/NF-kappaB pathway. ixazomib 125-133 nuclear factor kappa B subunit 1 Homo sapiens 55-64 33406639-10 2021 These results indicate that the activation of the SGK1/NF-kappaB pathway correlates with a low sensitivity to bortezomib and ixazomib, and a combination of bortezomib and ixazomib with an SGK1 or NF-kappaB inhibitor may be involved in the treatment of MM via activation of the SGK1/NF-kappaB pathway. ixazomib 171-179 serum/glucocorticoid regulated kinase 1 Homo sapiens 50-54 33406639-10 2021 These results indicate that the activation of the SGK1/NF-kappaB pathway correlates with a low sensitivity to bortezomib and ixazomib, and a combination of bortezomib and ixazomib with an SGK1 or NF-kappaB inhibitor may be involved in the treatment of MM via activation of the SGK1/NF-kappaB pathway. ixazomib 171-179 nuclear factor kappa B subunit 1 Homo sapiens 55-64 33406639-10 2021 These results indicate that the activation of the SGK1/NF-kappaB pathway correlates with a low sensitivity to bortezomib and ixazomib, and a combination of bortezomib and ixazomib with an SGK1 or NF-kappaB inhibitor may be involved in the treatment of MM via activation of the SGK1/NF-kappaB pathway. ixazomib 171-179 serum/glucocorticoid regulated kinase 1 Homo sapiens 188-192 33406639-10 2021 These results indicate that the activation of the SGK1/NF-kappaB pathway correlates with a low sensitivity to bortezomib and ixazomib, and a combination of bortezomib and ixazomib with an SGK1 or NF-kappaB inhibitor may be involved in the treatment of MM via activation of the SGK1/NF-kappaB pathway. ixazomib 171-179 nuclear factor kappa B subunit 1 Homo sapiens 196-205 33406639-10 2021 These results indicate that the activation of the SGK1/NF-kappaB pathway correlates with a low sensitivity to bortezomib and ixazomib, and a combination of bortezomib and ixazomib with an SGK1 or NF-kappaB inhibitor may be involved in the treatment of MM via activation of the SGK1/NF-kappaB pathway. ixazomib 171-179 serum/glucocorticoid regulated kinase 1 Homo sapiens 188-192 33406639-10 2021 These results indicate that the activation of the SGK1/NF-kappaB pathway correlates with a low sensitivity to bortezomib and ixazomib, and a combination of bortezomib and ixazomib with an SGK1 or NF-kappaB inhibitor may be involved in the treatment of MM via activation of the SGK1/NF-kappaB pathway. ixazomib 171-179 nuclear factor kappa B subunit 1 Homo sapiens 196-205 32931083-3 2020 Here, we report that the proteasome inhibitors bortezomib and ixazomib markedly increased protein levels of the osteoblastic key transcription factor osterix/Sp7 (Osx). ixazomib 62-70 Sp7 transcription factor Homo sapiens 150-161 32931083-3 2020 Here, we report that the proteasome inhibitors bortezomib and ixazomib markedly increased protein levels of the osteoblastic key transcription factor osterix/Sp7 (Osx). ixazomib 62-70 Sp7 transcription factor Homo sapiens 163-166 32653622-12 2020 B-cell activating factor (BAFF) plasma levels were significantly higher after ixazomib dosing in those who remained cGVHD-free compared to those developed cGVHD. ixazomib 78-86 TNF superfamily member 13b Homo sapiens 0-24 32653622-12 2020 B-cell activating factor (BAFF) plasma levels were significantly higher after ixazomib dosing in those who remained cGVHD-free compared to those developed cGVHD. ixazomib 78-86 TNF superfamily member 13b Homo sapiens 26-30 32350909-0 2020 Clinical benefit of ixazomib plus lenalidomide-dexamethasone in myeloma patients with non-canonical NF-kappaB pathway activation. ixazomib 20-28 nuclear factor kappa B subunit 1 Homo sapiens 100-109 32350909-6 2020 CONCLUSIONS: Adding ixazomib to Rd provides clinical benefit in MM tumors with increased non-canonical NF-kappaB pathway activity. ixazomib 20-28 nuclear factor kappa B subunit 1 Homo sapiens 103-112 32453773-6 2020 We have previously shown that ixazomib (IXA), a proteasome inhibitor used for treating multiple myeloma, effectively reduced the size of the infarct produced by global ischemia in isolated rat hearts and prevented degradation of the sarcoplasmic reticulum calcium release channel RyR2. ixazomib 30-38 ryanodine receptor 2 Rattus norvegicus 280-284 32019102-0 2020 Ixazomib Improves Bone Remodeling and Counteracts sonic Hedgehog signaling Inhibition Mediated by Myeloma Cells. ixazomib 0-8 sonic hedgehog signaling molecule Homo sapiens 50-64 32019102-7 2020 Given the key role of Sonic Hedgehog (SHH) signaling in bone homeostasis, we further investigated Ixazomib-induced SHH pathway activation. ixazomib 98-106 sonic hedgehog signaling molecule Homo sapiens 115-118 32019102-8 2020 This set of experiments showed that Ixazomib, but not Bortezomib, was able to bind the Smoothened (SMO) receptor leading to nuclear translocation of GLI1 in human MSCs. ixazomib 36-44 GLI family zinc finger 1 Homo sapiens 149-153 32019102-10 2020 In conclusion, our data demonstrated that Ixazomib regulates bone remodeling by decreasing osteoclastogenesis and prompting osteoblast differentiation via the canonical SHH signaling pathway activation, thus, representing a promising therapeutic option to improve the complex pathological condition of MM patients. ixazomib 42-50 sonic hedgehog signaling molecule Homo sapiens 169-172 31452195-0 2020 Combinatorial ixazomib and belinostat therapy induces NFE2L2-dependent apoptosis in Hodgkin and T-cell lymphoma. ixazomib 14-22 NFE2 like bZIP transcription factor 2 Homo sapiens 54-60 31452195-3 2020 In ixazomib-treated TCL and HL cells, transient inhibition followed by full recovery of proteasomal activity observed was accompanied by induction of proteasomal gene expression with NFE2L2 (also termed NRF2) as a prominent upstream regulator. ixazomib 3-11 NFE2 like bZIP transcription factor 2 Homo sapiens 183-189 31452195-3 2020 In ixazomib-treated TCL and HL cells, transient inhibition followed by full recovery of proteasomal activity observed was accompanied by induction of proteasomal gene expression with NFE2L2 (also termed NRF2) as a prominent upstream regulator. ixazomib 3-11 NFE2 like bZIP transcription factor 2 Homo sapiens 203-207 31452195-5 2020 In addition, CRISPR/Cas9 mediated knockdown of NFE2L2 in Jurkat cells resulted in a significant decrease in cell viability with ixazomib compared with untreated control cells. ixazomib 128-136 NFE2 like bZIP transcription factor 2 Homo sapiens 47-53 31452195-6 2020 Using transcriptomic and proteasomal activity evaluation of ixazomib, belinostat, or ixazomib + belinostat treated cells, we observed that NFE2L2, proteasome gene expression and functional recovery were abrogated by ixazomib + belinostat combination, resulting in synergistic drug activity in ixazomib-sensitive and -resistant cell lines and primary cells. ixazomib 60-68 NFE2 like bZIP transcription factor 2 Homo sapiens 139-145 31452195-6 2020 Using transcriptomic and proteasomal activity evaluation of ixazomib, belinostat, or ixazomib + belinostat treated cells, we observed that NFE2L2, proteasome gene expression and functional recovery were abrogated by ixazomib + belinostat combination, resulting in synergistic drug activity in ixazomib-sensitive and -resistant cell lines and primary cells. ixazomib 85-93 NFE2 like bZIP transcription factor 2 Homo sapiens 139-145 31452195-6 2020 Using transcriptomic and proteasomal activity evaluation of ixazomib, belinostat, or ixazomib + belinostat treated cells, we observed that NFE2L2, proteasome gene expression and functional recovery were abrogated by ixazomib + belinostat combination, resulting in synergistic drug activity in ixazomib-sensitive and -resistant cell lines and primary cells. ixazomib 85-93 NFE2 like bZIP transcription factor 2 Homo sapiens 139-145 31452195-6 2020 Using transcriptomic and proteasomal activity evaluation of ixazomib, belinostat, or ixazomib + belinostat treated cells, we observed that NFE2L2, proteasome gene expression and functional recovery were abrogated by ixazomib + belinostat combination, resulting in synergistic drug activity in ixazomib-sensitive and -resistant cell lines and primary cells. ixazomib 85-93 NFE2 like bZIP transcription factor 2 Homo sapiens 139-145 31452195-7 2020 Altogether, these results suggest that the synergistic activity of ixazomib + belinostat is mediated via inhibition NFE2L2-dependent proteasomal recovery and extended proteasomal inhibition culminating in increased cell death. ixazomib 67-75 NFE2 like bZIP transcription factor 2 Homo sapiens 116-122 31540997-5 2019 Herein we show that bortezomib and next-generation proteasome inhibitors ixazomib and carfilzomib markedly induce AIRAP expression in human melanoma at concentrations comparable to plasma-levels in treated patients. ixazomib 73-81 zinc finger AN1-type containing 2A Homo sapiens 114-119 31273172-9 2019 Patients with an estimated glomerular filtration rate of less than 50mL/min/1.73m2 received a lower cumulative dose of ixazomib and lenalidomide than those with other rates. ixazomib 119-127 CD59 molecule (CD59 blood group) Homo sapiens 72-77 30487243-9 2019 Mechanistically, ixazomib treatment resulted in the accumulation of polyubiquitinated proteins, including phosphorylated IkappaBalpha with a significant reduction of p65 subunit nuclear translocation. ixazomib 17-25 NFKB inhibitor alpha Homo sapiens 121-133 30487243-9 2019 Mechanistically, ixazomib treatment resulted in the accumulation of polyubiquitinated proteins, including phosphorylated IkappaBalpha with a significant reduction of p65 subunit nuclear translocation. ixazomib 17-25 RELA proto-oncogene, NF-kB subunit Homo sapiens 166-169 30487243-11 2019 IMPLICATIONS: This preclinical study supports the use of oral proteasome inhibitor ixazomib for targeting NF-kappaB signaling in the treatment of EBV-associated B-cell neoplasms.Visual Overview: http://mcr.aacrjournals.org/content/molcanres/17/4/839/F1.large.jpg. ixazomib 83-91 nuclear factor kappa B subunit 1 Homo sapiens 106-115 30902965-8 2019 MLN9708, a proteasome inhibitor, potently induced autophagy via the induction of MAP1LC3B and sensitized the cell to autophagy-mediated cell death in VHL-deficient and VHL-mutant (L101A) cells. ixazomib 0-7 microtubule-associated protein 1 light chain 3 beta Mus musculus 81-89 30359552-0 2019 Ixazomib promotes CHOP-dependent DR5 induction and apoptosis in colorectal cancer cells. ixazomib 0-8 DNA damage inducible transcript 3 Homo sapiens 18-22 30359552-0 2019 Ixazomib promotes CHOP-dependent DR5 induction and apoptosis in colorectal cancer cells. ixazomib 0-8 TNF receptor superfamily member 10b Homo sapiens 33-36 30359552-4 2019 Western blotting and real-time RT-PCR were performed to detect ixazomib-induced DR5 upregulation. ixazomib 63-71 TNF receptor superfamily member 10b Homo sapiens 80-83 30359552-8 2019 Our findings indicated that ixazomib treatment induces CHOP-dependent DR5 induction, irrespective of p53 status. ixazomib 28-36 DNA damage inducible transcript 3 Homo sapiens 55-59 30359552-8 2019 Our findings indicated that ixazomib treatment induces CHOP-dependent DR5 induction, irrespective of p53 status. ixazomib 28-36 TNF receptor superfamily member 10b Homo sapiens 70-73 30359552-9 2019 Furthermore, DR5 is necessary for ixazomib-mediated apoptosis. ixazomib 34-42 TNF receptor superfamily member 10b Homo sapiens 13-16 30359552-10 2019 Ixazomib also synergized with TRAIL to induce marked apoptosis via DR5 in CRC cells. ixazomib 0-8 TNF receptor superfamily member 10b Homo sapiens 67-70 30359552-11 2019 CONCLUSIONS: Our findings further suggested that ixazomib sensitizes TRAIL/death receptor signaling pathway-targeted CRC and suggested that DR5 induction could be a valuable indicator of ixazomib sensitivity. ixazomib 49-57 TNF superfamily member 10 Homo sapiens 69-74 30359552-11 2019 CONCLUSIONS: Our findings further suggested that ixazomib sensitizes TRAIL/death receptor signaling pathway-targeted CRC and suggested that DR5 induction could be a valuable indicator of ixazomib sensitivity. ixazomib 187-195 TNF receptor superfamily member 10b Homo sapiens 140-143 30089730-9 2018 Finally, we performed proof-of-principle experiments using a currently approved proteasome inhibitor, ixazomib, to target FOXM1 and demonstrated a therapeutic response in AML patient samples and animal models of AML that correlates with the suppression of FOXM1 and its transcriptional targets. ixazomib 102-110 forkhead box M1 Homo sapiens 122-127 30089730-9 2018 Finally, we performed proof-of-principle experiments using a currently approved proteasome inhibitor, ixazomib, to target FOXM1 and demonstrated a therapeutic response in AML patient samples and animal models of AML that correlates with the suppression of FOXM1 and its transcriptional targets. ixazomib 102-110 forkhead box M1 Homo sapiens 256-261 29699990-12 2018 Finally, a potential therapeutic application of proteasome inhibitors (bortezomib and ixazomib), which are known to inhibit NF-kappaB, was identified through their ability to decrease proliferation and increase apoptosis in LGL leukemia cell lines and primary patient cells. ixazomib 86-94 nuclear factor kappa B subunit 1 Homo sapiens 124-133 29643240-6 2018 Ixazomib, a proteasome inhibitor in clinical use, blocks this pathway, increasing the abundance of Casp8p41 and causing more cells to die in a Casp8p41-dependent manner.IMPORTANCE The Casp8p41 pathway of cell death is unique to HIV-infected cells yet is blocked by Bcl2. ixazomib 0-8 caspase 8 Homo sapiens 99-105 29643240-6 2018 Ixazomib, a proteasome inhibitor in clinical use, blocks this pathway, increasing the abundance of Casp8p41 and causing more cells to die in a Casp8p41-dependent manner.IMPORTANCE The Casp8p41 pathway of cell death is unique to HIV-infected cells yet is blocked by Bcl2. ixazomib 0-8 BCL2 apoptosis regulator Homo sapiens 265-269 28800141-0 2018 Effects of Strong CYP3A Inhibition and Induction on the Pharmacokinetics of Ixazomib, an Oral Proteasome Inhibitor: Results of Drug-Drug Interaction Studies in Patients With Advanced Solid Tumors or Lymphoma and a Physiologically Based Pharmacokinetic Analysis. ixazomib 76-84 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 18-23 28800141-2 2018 However, at higher than clinical concentrations, ixazomib was metabolized by multiple CYP isoforms, with the estimated relative contribution being highest for CYP3A at 42%. ixazomib 49-57 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 159-164 28800141-9 2018 The clinical drug-drug interaction study results were reconciled well by a physiologically based pharmacokinetic model that incorporated a minor contribution of CYP3A to overall ixazomib clearance and quantitatively considered the strength of induction of CYP3A and intestinal P-glycoprotein by rifampin. ixazomib 178-186 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 161-166 28800141-10 2018 On the basis of these study results, the ixazomib prescribing information recommends that patients should avoid concomitant administration of strong CYP3A inducers with ixazomib. ixazomib 41-49 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 149-154 28800141-10 2018 On the basis of these study results, the ixazomib prescribing information recommends that patients should avoid concomitant administration of strong CYP3A inducers with ixazomib. ixazomib 169-177 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 149-154 28842936-4 2017 Significant reductions in cell viability, NF-kappaB activation, and GATA-3 expression were observed preclinically in ixazomib-treated cells. ixazomib 117-125 GATA binding protein 3 Homo sapiens 68-74 28842936-6 2017 Concordant with our preclinical observations, a significant reduction in NF-kappaB activation and GATA-3 expression was observed in an exceptional responder following one month of treatment with ixazomib. ixazomib 195-203 GATA binding protein 3 Homo sapiens 98-104 28842936-7 2017 While ixazomib had limited activity in this small and heterogeneous cohort of patients, inhibition of the NF-kappaB/GATA-3 axis in a single exceptional responder suggests that ixazomib may have utility in appropriately selected patients or in combination with other agents. ixazomib 176-184 GATA binding protein 3 Homo sapiens 116-122 29416618-5 2018 In addition, we showed that ixazomib induces apoptosis and the DNA damage response pathway, through activation of the checkpoint kinase 2 (CHK2). ixazomib 28-36 checkpoint kinase 2 Homo sapiens 118-137 29416618-5 2018 In addition, we showed that ixazomib induces apoptosis and the DNA damage response pathway, through activation of the checkpoint kinase 2 (CHK2). ixazomib 28-36 checkpoint kinase 2 Homo sapiens 139-143 29416618-6 2018 Hence, pharmacological inhibition of CHK2 enhances the anti-tumor activity of ixazomib in DLBCL cells. ixazomib 78-86 checkpoint kinase 2 Homo sapiens 37-41 28790851-13 2017 It exerted anticancer effects through targeting the expression of DEGs, such as HSPA6, APCDD1, TP53, and JUN, and affecting the signaling pathways including apoptosis and cell cycle pathway, which demonstrated the promising potential of ixazomib for CRC therapy. ixazomib 237-245 heat shock protein family A (Hsp70) member 6 Homo sapiens 80-85 28790851-13 2017 It exerted anticancer effects through targeting the expression of DEGs, such as HSPA6, APCDD1, TP53, and JUN, and affecting the signaling pathways including apoptosis and cell cycle pathway, which demonstrated the promising potential of ixazomib for CRC therapy. ixazomib 237-245 APC down-regulated 1 Homo sapiens 87-93 28790851-13 2017 It exerted anticancer effects through targeting the expression of DEGs, such as HSPA6, APCDD1, TP53, and JUN, and affecting the signaling pathways including apoptosis and cell cycle pathway, which demonstrated the promising potential of ixazomib for CRC therapy. ixazomib 237-245 tumor protein p53 Homo sapiens 95-99 28495797-0 2017 Ixazomib enhances parathyroid hormone-induced beta-catenin/T-cell factor signaling by dissociating beta-catenin from the parathyroid hormone receptor. ixazomib 0-8 hepatocyte nuclear factor 4 alpha Homo sapiens 46-72 28495797-0 2017 Ixazomib enhances parathyroid hormone-induced beta-catenin/T-cell factor signaling by dissociating beta-catenin from the parathyroid hormone receptor. ixazomib 0-8 catenin beta 1 Homo sapiens 46-58 27783987-9 2016 However, the combination of ixazomib with IFN-alpha significantly enhanced ixazomib"s ability to induce apoptotic cell death in BRAF V600E mutant and BRAF wild-type human melanoma tumor cells. ixazomib 28-36 B-Raf proto-oncogene, serine/threonine kinase Homo sapiens 128-132 27783987-9 2016 However, the combination of ixazomib with IFN-alpha significantly enhanced ixazomib"s ability to induce apoptotic cell death in BRAF V600E mutant and BRAF wild-type human melanoma tumor cells. ixazomib 28-36 B-Raf proto-oncogene, serine/threonine kinase Homo sapiens 150-154 27783987-9 2016 However, the combination of ixazomib with IFN-alpha significantly enhanced ixazomib"s ability to induce apoptotic cell death in BRAF V600E mutant and BRAF wild-type human melanoma tumor cells. ixazomib 75-83 interferon alpha 1 Homo sapiens 42-51 27783987-9 2016 However, the combination of ixazomib with IFN-alpha significantly enhanced ixazomib"s ability to induce apoptotic cell death in BRAF V600E mutant and BRAF wild-type human melanoma tumor cells. ixazomib 75-83 B-Raf proto-oncogene, serine/threonine kinase Homo sapiens 128-132 27783987-9 2016 However, the combination of ixazomib with IFN-alpha significantly enhanced ixazomib"s ability to induce apoptotic cell death in BRAF V600E mutant and BRAF wild-type human melanoma tumor cells. ixazomib 75-83 B-Raf proto-oncogene, serine/threonine kinase Homo sapiens 150-154 27783987-10 2016 The combination of ixazomib and IFN-alpha also enhanced inhibition of cell proliferation in BRAF V600E mutant melanoma tumor cells; however, this was not seen in BRAF wild-type cells. ixazomib 19-27 B-Raf proto-oncogene, serine/threonine kinase Homo sapiens 92-96 27783987-11 2016 Ixazomib-induced apoptosis was associated with processing of the pro-apoptotic proteins procaspase-3, -7, -8, and -9, and cleavage of poly-ADP-ribose polymerase (PARP). ixazomib 0-8 caspase 3 Homo sapiens 88-116 27783987-11 2016 Ixazomib-induced apoptosis was associated with processing of the pro-apoptotic proteins procaspase-3, -7, -8, and -9, and cleavage of poly-ADP-ribose polymerase (PARP). ixazomib 0-8 poly(ADP-ribose) polymerase 1 Homo sapiens 134-160 27783987-11 2016 Ixazomib-induced apoptosis was associated with processing of the pro-apoptotic proteins procaspase-3, -7, -8, and -9, and cleavage of poly-ADP-ribose polymerase (PARP). ixazomib 0-8 poly(ADP-ribose) polymerase 1 Homo sapiens 162-166 27783987-12 2016 In an in vivo xenograft model of human melanoma, combination treatment with IFN-alpha-2b and ixazomib demonstrated a significant reduction in tumor volume when compared to vehicle (p = 0.005) and single therapy ixazomib (p = 0.017) and IFN-alpha-2b (p = 0.036). ixazomib 93-101 interferon alpha 1 Homo sapiens 236-245 27783987-12 2016 In an in vivo xenograft model of human melanoma, combination treatment with IFN-alpha-2b and ixazomib demonstrated a significant reduction in tumor volume when compared to vehicle (p = 0.005) and single therapy ixazomib (p = 0.017) and IFN-alpha-2b (p = 0.036). ixazomib 211-219 interferon alpha 1 Homo sapiens 76-85 27783987-13 2016 These pre-clinical results support further evaluation of combination treatment with ixazomib and IFN-alpha for the treatment of advanced BRAF V600E mutant melanoma. ixazomib 84-92 B-Raf proto-oncogene, serine/threonine kinase Homo sapiens 137-141 27325500-3 2016 When investigating the proteasome inhibitors bortezomib, carfilzomib, and ixazomib, we observed increased luminescence for bortezomib and ixazomib, but not for carfilzomib, in a pregnane-X-receptor (PXR) reporter gene assay, which was inconsistent with the mRNA expression levels of the main PXR target gene CYP3A4. ixazomib 74-82 nuclear receptor subfamily 1 group I member 2 Homo sapiens 178-197 27325500-3 2016 When investigating the proteasome inhibitors bortezomib, carfilzomib, and ixazomib, we observed increased luminescence for bortezomib and ixazomib, but not for carfilzomib, in a pregnane-X-receptor (PXR) reporter gene assay, which was inconsistent with the mRNA expression levels of the main PXR target gene CYP3A4. ixazomib 74-82 nuclear receptor subfamily 1 group I member 2 Homo sapiens 199-202 27325500-3 2016 When investigating the proteasome inhibitors bortezomib, carfilzomib, and ixazomib, we observed increased luminescence for bortezomib and ixazomib, but not for carfilzomib, in a pregnane-X-receptor (PXR) reporter gene assay, which was inconsistent with the mRNA expression levels of the main PXR target gene CYP3A4. ixazomib 74-82 nuclear receptor subfamily 1 group I member 2 Homo sapiens 292-295 27325500-3 2016 When investigating the proteasome inhibitors bortezomib, carfilzomib, and ixazomib, we observed increased luminescence for bortezomib and ixazomib, but not for carfilzomib, in a pregnane-X-receptor (PXR) reporter gene assay, which was inconsistent with the mRNA expression levels of the main PXR target gene CYP3A4. ixazomib 74-82 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 308-314 27325500-3 2016 When investigating the proteasome inhibitors bortezomib, carfilzomib, and ixazomib, we observed increased luminescence for bortezomib and ixazomib, but not for carfilzomib, in a pregnane-X-receptor (PXR) reporter gene assay, which was inconsistent with the mRNA expression levels of the main PXR target gene CYP3A4. ixazomib 138-146 nuclear receptor subfamily 1 group I member 2 Homo sapiens 178-197 27325500-3 2016 When investigating the proteasome inhibitors bortezomib, carfilzomib, and ixazomib, we observed increased luminescence for bortezomib and ixazomib, but not for carfilzomib, in a pregnane-X-receptor (PXR) reporter gene assay, which was inconsistent with the mRNA expression levels of the main PXR target gene CYP3A4. ixazomib 138-146 nuclear receptor subfamily 1 group I member 2 Homo sapiens 199-202 27325500-3 2016 When investigating the proteasome inhibitors bortezomib, carfilzomib, and ixazomib, we observed increased luminescence for bortezomib and ixazomib, but not for carfilzomib, in a pregnane-X-receptor (PXR) reporter gene assay, which was inconsistent with the mRNA expression levels of the main PXR target gene CYP3A4. ixazomib 138-146 nuclear receptor subfamily 1 group I member 2 Homo sapiens 292-295 27325500-3 2016 When investigating the proteasome inhibitors bortezomib, carfilzomib, and ixazomib, we observed increased luminescence for bortezomib and ixazomib, but not for carfilzomib, in a pregnane-X-receptor (PXR) reporter gene assay, which was inconsistent with the mRNA expression levels of the main PXR target gene CYP3A4. ixazomib 138-146 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 308-314 27529620-9 2016 Ixazomib also prevented the 50% reduction in RyR2 content observed after ischemia. ixazomib 0-8 ryanodine receptor 2 Rattus norvegicus 45-49 27429848-0 2016 BH3 mimetic ABT-737 sensitizes colorectal cancer cells to ixazomib through MCL-1 downregulation and autophagy inhibition. ixazomib 58-66 MCL1 apoptosis regulator, BCL2 family member Homo sapiens 75-80 26988986-0 2016 Proteasomal Inhibition by Ixazomib Induces CHK1 and MYC-Dependent Cell Death in T-cell and Hodgkin Lymphoma. ixazomib 26-34 checkpoint kinase 1 Homo sapiens 43-47 26988986-7 2016 The predicted activity for tumor suppressors and oncogenes, the impact on "hallmarks of cancer," and the analysis of key significant genes from global transcriptome analysis for ixazomib strongly favored tumor inhibition via downregulation of MYC and CHK1, its target genes. ixazomib 178-186 MYC proto-oncogene, bHLH transcription factor Homo sapiens 243-246 26988986-7 2016 The predicted activity for tumor suppressors and oncogenes, the impact on "hallmarks of cancer," and the analysis of key significant genes from global transcriptome analysis for ixazomib strongly favored tumor inhibition via downregulation of MYC and CHK1, its target genes. ixazomib 178-186 checkpoint kinase 1 Homo sapiens 251-255 26988986-8 2016 Furthermore, in ixazomib-treated lymphoma cells, we identified that CHK1 was involved in the regulation of MYC expression through chromatin modification involving histone H3 acetylation via chromatin immunoprecipitation. ixazomib 16-24 checkpoint kinase 1 Homo sapiens 68-72 26988986-8 2016 Furthermore, in ixazomib-treated lymphoma cells, we identified that CHK1 was involved in the regulation of MYC expression through chromatin modification involving histone H3 acetylation via chromatin immunoprecipitation. ixazomib 16-24 MYC proto-oncogene, bHLH transcription factor Homo sapiens 107-110 26988986-10 2016 Altogether, ixazomib significantly downregulates MYC and induces potent cell death in T-cell lymphoma and Hodgkin lymphoma, and we identified that combinatorial therapy with anti-CHK1 treatment represents a rational and novel therapeutic approach. ixazomib 12-20 MYC proto-oncogene, bHLH transcription factor Homo sapiens 49-52 26988986-10 2016 Altogether, ixazomib significantly downregulates MYC and induces potent cell death in T-cell lymphoma and Hodgkin lymphoma, and we identified that combinatorial therapy with anti-CHK1 treatment represents a rational and novel therapeutic approach. ixazomib 12-20 checkpoint kinase 1 Homo sapiens 179-183 27217076-6 2016 MLN9708 also enhanced Dox-induced JNK and p38 phosphorylation and inhibited Dox-induced IkappaBalpha degradation. ixazomib 0-7 mitogen-activated protein kinase 14 Homo sapiens 42-45 27217076-6 2016 MLN9708 also enhanced Dox-induced JNK and p38 phosphorylation and inhibited Dox-induced IkappaBalpha degradation. ixazomib 0-7 NFKB inhibitor alpha Homo sapiens 88-100 26634271-0 2016 Selective Toxicity of Investigational Ixazomib for Human Leukemia Cells Expressing Mutant Cytoplasmic NPM1: Role of Reactive Oxygen Species. ixazomib 38-46 nucleophosmin 1 Homo sapiens 102-106 26634271-4 2016 RESULTS: Apoptosis induced by ixazomib was abrogated by knockdown of NPM1/NPMc(+)expression using an inducible shRNA construct and enhanced by NPMc(+)overexpression. ixazomib 30-38 nucleophosmin 1 Homo sapiens 69-73 26709701-0 2015 KRAS Genotype Correlates with Proteasome Inhibitor Ixazomib Activity in Preclinical In Vivo Models of Colon and Non-Small Cell Lung Cancer: Potential Role of Tumor Metabolism. ixazomib 51-59 KRAS proto-oncogene, GTPase Homo sapiens 0-4 26709701-4 2015 A survey of 14 non-small cell lung cancer (NSCLC) and 6 colon xenografts showed a striking relationship between ixazomib activity and KRAS genotype; tumors with wild-type (WT) KRAS were more sensitive to ixazomib than tumors harboring KRAS activating mutations. ixazomib 112-120 KRAS proto-oncogene, GTPase Homo sapiens 134-138 26709701-4 2015 A survey of 14 non-small cell lung cancer (NSCLC) and 6 colon xenografts showed a striking relationship between ixazomib activity and KRAS genotype; tumors with wild-type (WT) KRAS were more sensitive to ixazomib than tumors harboring KRAS activating mutations. ixazomib 112-120 KRAS proto-oncogene, GTPase Homo sapiens 176-180 26709701-4 2015 A survey of 14 non-small cell lung cancer (NSCLC) and 6 colon xenografts showed a striking relationship between ixazomib activity and KRAS genotype; tumors with wild-type (WT) KRAS were more sensitive to ixazomib than tumors harboring KRAS activating mutations. ixazomib 112-120 KRAS proto-oncogene, GTPase Homo sapiens 176-180 26709701-5 2015 To confirm the association between KRAS genotype and ixazomib sensitivity, we used SW48 isogenic colon cancer cell lines. ixazomib 53-61 KRAS proto-oncogene, GTPase Homo sapiens 35-39 26709701-7 2015 SW48 KRAS WT tumors, but neither SW48-KRAS-G13D tumors nor SW48-KRAS-G12V tumors, were sensitive to ixazomib in vivo. ixazomib 100-108 KRAS proto-oncogene, GTPase Homo sapiens 5-9 26709701-13 2015 The non-clinical data presented here show a correlation between KRAS genotype and ixazomib sensitivity in NSCLC and colon xenografts and provide new evidence of regulation of key metabolic pathways by proteasome inhibition. ixazomib 82-90 KRAS proto-oncogene, GTPase Homo sapiens 64-68 26141494-3 2015 METHODS: Nonclinical studies assessed (1) the in vitro binding of ixazomib to the hERG channel and (2) its effect on QT/QTc in dogs (N = 4) via telemetry. ixazomib 66-74 ETS transcription factor ERG Homo sapiens 82-86 26141494-6 2015 RESULTS: In vitro binding potency for ixazomib to the hERG channel was weak (K i 24.9 muM; IC50 59.6 muM), and nonclinical telemetry studies showed no QT/QTc prolongation at doses up to 4.2 mg/m(2). ixazomib 38-46 ETS transcription factor ERG Homo sapiens 54-58 26141494-6 2015 RESULTS: In vitro binding potency for ixazomib to the hERG channel was weak (K i 24.9 muM; IC50 59.6 muM), and nonclinical telemetry studies showed no QT/QTc prolongation at doses up to 4.2 mg/m(2). ixazomib 38-46 latexin Homo sapiens 86-89 26141494-6 2015 RESULTS: In vitro binding potency for ixazomib to the hERG channel was weak (K i 24.9 muM; IC50 59.6 muM), and nonclinical telemetry studies showed no QT/QTc prolongation at doses up to 4.2 mg/m(2). ixazomib 38-46 latexin Homo sapiens 101-104 25919767-0 2015 Calcineurin Inhibitor Minimization With Ixazomib, an Investigational Proteasome Inhibitor, for the Prevention of Antibody Mediated Rejection in a Preclinical Model. ixazomib 40-48 calcineurin binding protein 1 Rattus norvegicus 0-21 25919767-5 2015 RESULTS: Compared to untreated animals, ixazomib alone or in combination with 1/2 dose CsA reduced donor-specific antibody, intragraft transcripts for chemokines CCL-21 and CXCL-13, and CD19 expression in both sensitized and naive transplants. ixazomib 40-48 C-C motif chemokine ligand 21 Rattus norvegicus 162-168 25919767-5 2015 RESULTS: Compared to untreated animals, ixazomib alone or in combination with 1/2 dose CsA reduced donor-specific antibody, intragraft transcripts for chemokines CCL-21 and CXCL-13, and CD19 expression in both sensitized and naive transplants. ixazomib 40-48 C-X-C motif chemokine ligand 13 Rattus norvegicus 173-180 25919767-5 2015 RESULTS: Compared to untreated animals, ixazomib alone or in combination with 1/2 dose CsA reduced donor-specific antibody, intragraft transcripts for chemokines CCL-21 and CXCL-13, and CD19 expression in both sensitized and naive transplants. ixazomib 40-48 CD19 molecule Rattus norvegicus 186-190 26099967-3 2015 All established PI (bortezomib and carfilzomib), as well as experimental drugs in the field (dalanzomib, oprozomib, and ixazomib), by design target the rate-limiting beta5 subunit. ixazomib 120-128 adaptor related protein complex 5 subunit beta 1 Homo sapiens 166-171 25234165-5 2015 MiR-29b was significantly reduced in bortezomib-resistant cells as well as in cells resistant to second-generation PIs carfilzomib and ixazomib. ixazomib 135-143 microRNA 29b-1 Homo sapiens 0-7 24120758-4 2014 Here, we show that similar to bortezomib, the novel proteasome inhibitors carfilzomib and ixazomib, as well as staurosporine and adaphostin, induce the generation of Mcl-1(128-350) in MM cells. ixazomib 90-98 MCL1 apoptosis regulator, BCL2 family member Homo sapiens 166-171 24292417-0 2013 (18)F-FDG-PET/CT imaging in an IL-6- and MYC-driven mouse model of human multiple myeloma affords objective evaluation of plasma cell tumor progression and therapeutic response to the proteasome inhibitor ixazomib. ixazomib 205-213 interleukin 6 Mus musculus 31-44 24285522-0 2013 Syntheses of C-13 and C-14-labeled versions of the investigational proteasome inhibitor MLN9708. ixazomib 88-95 homeobox C13 Homo sapiens 13-17