Skip to main content
Top
Published in: Journal of Experimental & Clinical Cancer Research 1/2018

Open Access 01-12-2018 | Research

FBP1 loss contributes to BET inhibitors resistance by undermining c-Myc expression in pancreatic ductal adenocarcinoma

Authors: Bo Wang, Ping Fan, Jingyuan Zhao, Heyu Wu, Xin Jin, Heshui Wu

Published in: Journal of Experimental & Clinical Cancer Research | Issue 1/2018

Login to get access

Abstract

Background

Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal tumor types worldwide. BET inhibitors display anti-tumor activity in pancreatic cancer, however the cells often develop resistance after a long-term treatment and the underlying molecular basis is not fully understood.

Methods

Drug screening assay in Fructose-1, 6-biphosphatase (FBP1) knockdown or overexpressing pancreatic cancer cells was performed. Tumor cell motility, FBP1 protein and mRNA changes were investigated after BET inhibitors treatment. The interaction between TRIM28 and FBP1 after BET inhibitors treatment was examined by Co-immunoprecipitation (IP) and GST pull-down. The relationship between FBP1 and c-Myc was examined by western blot, RT-qPCR and immunohistochemistry (IHC).

Results

The expression of FBP1 protein increased the sensitivity of pancreatic cancer cells to JQ1. Furthermore, we showed that JQ1 stabilized FBP1 protein level by disrupting the interaction between FBP1 and TRIM28 in pancreatic cancer cells. Moreover, we demonstrated that FBP1 promoted c-Myc degradation through disrupting the ERK-c-Myc axis.

Conclusions

FBP1 modulates the sensitivity of pancreatic cancer cells to BET inhibitors by decreasing the expression of c-Myc. These findings highlight FBP1 could be used as a therapeutic niche for patient-tailored therapies.
Appendix
Available only for authorised users
Literature
2.
go back to reference Von Hoff DD, Ervin T, Arena FP, Chiorean EG, Infante J, Moore M, Seay T, Tjulandin SA, Ma WW, Saleh MN, et al. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N Engl J Med. 2013;369:1691–703.CrossRefPubMedPubMedCentral Von Hoff DD, Ervin T, Arena FP, Chiorean EG, Infante J, Moore M, Seay T, Tjulandin SA, Ma WW, Saleh MN, et al. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N Engl J Med. 2013;369:1691–703.CrossRefPubMedPubMedCentral
3.
go back to reference Rathert P, Roth M, Neumann T, Muerdter F, Roe JS, Muhar M, Deswal S, Cerny-Reiterer S, Peter B, Jude J, et al. Transcriptional plasticity promotes primary and acquired resistance to BET inhibition. Nature. 2015;525:543–7.CrossRefPubMedPubMedCentral Rathert P, Roth M, Neumann T, Muerdter F, Roe JS, Muhar M, Deswal S, Cerny-Reiterer S, Peter B, Jude J, et al. Transcriptional plasticity promotes primary and acquired resistance to BET inhibition. Nature. 2015;525:543–7.CrossRefPubMedPubMedCentral
4.
go back to reference Mazur PK, Herner A, Mello SS, Wirth M, Hausmann S, Sanchez-Rivera FJ, Lofgren SM, Kuschma T, Hahn SA, Vangala D, et al. Combined inhibition of BET family proteins and histone deacetylases as a potential epigenetics-based therapy for pancreatic ductal adenocarcinoma. Nat Med. 2015;21:1163–71.CrossRefPubMedPubMedCentral Mazur PK, Herner A, Mello SS, Wirth M, Hausmann S, Sanchez-Rivera FJ, Lofgren SM, Kuschma T, Hahn SA, Vangala D, et al. Combined inhibition of BET family proteins and histone deacetylases as a potential epigenetics-based therapy for pancreatic ductal adenocarcinoma. Nat Med. 2015;21:1163–71.CrossRefPubMedPubMedCentral
5.
go back to reference Lenhart R, Kirov S, Desilva H, Cao J, Lei M, Johnston K, Peterson R, Schweizer L, Purandare A, Ross-Macdonald P, et al. Sensitivity of small cell lung Cancer to BET inhibition is mediated by regulation of ASCL1 gene expression. Mol Cancer Ther. 2015;14:2167–74.CrossRefPubMed Lenhart R, Kirov S, Desilva H, Cao J, Lei M, Johnston K, Peterson R, Schweizer L, Purandare A, Ross-Macdonald P, et al. Sensitivity of small cell lung Cancer to BET inhibition is mediated by regulation of ASCL1 gene expression. Mol Cancer Ther. 2015;14:2167–74.CrossRefPubMed
6.
go back to reference Blee AM, Liu S, Wang L, Huang H. BET bromodomain-mediated interaction between ERG and BRD4 promotes prostate cancer cell invasion. Oncotarget. 2016;7:38319–32.CrossRefPubMedPubMedCentral Blee AM, Liu S, Wang L, Huang H. BET bromodomain-mediated interaction between ERG and BRD4 promotes prostate cancer cell invasion. Oncotarget. 2016;7:38319–32.CrossRefPubMedPubMedCentral
7.
go back to reference Sahai V, Kumar K, Knab LM, Chow CR, Raza SS, Bentrem DJ, Ebine K, Munshi HG. BET bromodomain inhibitors block growth of pancreatic cancer cells in three-dimensional collagen. Mol Cancer Ther. 2014;13:1907–17.CrossRefPubMedPubMedCentral Sahai V, Kumar K, Knab LM, Chow CR, Raza SS, Bentrem DJ, Ebine K, Munshi HG. BET bromodomain inhibitors block growth of pancreatic cancer cells in three-dimensional collagen. Mol Cancer Ther. 2014;13:1907–17.CrossRefPubMedPubMedCentral
8.
go back to reference Andricovich J, Perkail S, Kai Y, Casasanta N, Peng W, Tzatsos A. Loss of KDM6A activates super-enhancers to induce gender-specific squamous-like pancreatic Cancer and confers sensitivity to BET inhibitors. Cancer Cell. 2018;33:512–26. e518CrossRefPubMed Andricovich J, Perkail S, Kai Y, Casasanta N, Peng W, Tzatsos A. Loss of KDM6A activates super-enhancers to induce gender-specific squamous-like pancreatic Cancer and confers sensitivity to BET inhibitors. Cancer Cell. 2018;33:512–26. e518CrossRefPubMed
9.
go back to reference Bian B, Bigonnet M, Gayet O, Loncle C, Maignan A, Gilabert M, Moutardier V, Garcia S, Turrini O, Delpero JR, et al. Gene expression profiling of patient-derived pancreatic cancer xenografts predicts sensitivity to the BET bromodomain inhibitor JQ1: implications for individualized medicine efforts. EMBO Mol Med. 2017;9:482–97.CrossRefPubMedPubMedCentral Bian B, Bigonnet M, Gayet O, Loncle C, Maignan A, Gilabert M, Moutardier V, Garcia S, Turrini O, Delpero JR, et al. Gene expression profiling of patient-derived pancreatic cancer xenografts predicts sensitivity to the BET bromodomain inhibitor JQ1: implications for individualized medicine efforts. EMBO Mol Med. 2017;9:482–97.CrossRefPubMedPubMedCentral
10.
go back to reference Kumar K, Raza SS, Knab LM, Chow CR, Kwok B, Bentrem DJ, Popovic R, Ebine K, Licht JD, Munshi HG. GLI2-dependent c-MYC upregulation mediates resistance of pancreatic cancer cells to the BET bromodomain inhibitor JQ1. Sci Rep. 2015;5:9489.CrossRefPubMedPubMedCentral Kumar K, Raza SS, Knab LM, Chow CR, Kwok B, Bentrem DJ, Popovic R, Ebine K, Licht JD, Munshi HG. GLI2-dependent c-MYC upregulation mediates resistance of pancreatic cancer cells to the BET bromodomain inhibitor JQ1. Sci Rep. 2015;5:9489.CrossRefPubMedPubMedCentral
11.
go back to reference Wirth M, Mahboobi S, Kramer OH, Schneider G. Concepts to target MYC in pancreatic Cancer. Mol Cancer Ther. 2016;15:1792–8.CrossRefPubMed Wirth M, Mahboobi S, Kramer OH, Schneider G. Concepts to target MYC in pancreatic Cancer. Mol Cancer Ther. 2016;15:1792–8.CrossRefPubMed
12.
go back to reference Kortlever RM, Sodir NM, Wilson CH, Burkhart DL, Pellegrinet L, Brown Swigart L, Littlewood TD, Evan GI. Myc cooperates with Ras by programming inflammation and immune suppression. Cell. 2017;171:1301–15. e1314CrossRefPubMedPubMedCentral Kortlever RM, Sodir NM, Wilson CH, Burkhart DL, Pellegrinet L, Brown Swigart L, Littlewood TD, Evan GI. Myc cooperates with Ras by programming inflammation and immune suppression. Cell. 2017;171:1301–15. e1314CrossRefPubMedPubMedCentral
13.
go back to reference Xin B, Yamamoto M, Fujii K, Ooshio T, Chen X, Okada Y, Watanabe K, Miyokawa N, Furukawa H, Nishikawa Y. Critical role of Myc activation in mouse hepatocarcinogenesis induced by the activation of AKT and RAS pathways. Oncogene. 2017;36:5087–97.CrossRefPubMedPubMedCentral Xin B, Yamamoto M, Fujii K, Ooshio T, Chen X, Okada Y, Watanabe K, Miyokawa N, Furukawa H, Nishikawa Y. Critical role of Myc activation in mouse hepatocarcinogenesis induced by the activation of AKT and RAS pathways. Oncogene. 2017;36:5087–97.CrossRefPubMedPubMedCentral
14.
go back to reference Jiang SH, Li J, Dong FY, Yang JY, Liu DJ, Yang XM, Wang YH, Yang MW, Fu XL, Zhang XX, et al. Increased serotonin signaling contributes to the Warburg effect in pancreatic tumor cells under metabolic stress and promotes growth of pancreatic tumors in mice. Gastroenterology. 2017;153:277–91. e219CrossRefPubMed Jiang SH, Li J, Dong FY, Yang JY, Liu DJ, Yang XM, Wang YH, Yang MW, Fu XL, Zhang XX, et al. Increased serotonin signaling contributes to the Warburg effect in pancreatic tumor cells under metabolic stress and promotes growth of pancreatic tumors in mice. Gastroenterology. 2017;153:277–91. e219CrossRefPubMed
15.
go back to reference Li B, Qiu B, Lee DS, Walton ZE, Ochocki JD, Mathew LK, Mancuso A, Gade TP, Keith B, Nissim I, Simon MC. Fructose-1,6-bisphosphatase opposes renal carcinoma progression. Nature. 2014;513:251–5.CrossRefPubMedPubMedCentral Li B, Qiu B, Lee DS, Walton ZE, Ochocki JD, Mathew LK, Mancuso A, Gade TP, Keith B, Nissim I, Simon MC. Fructose-1,6-bisphosphatase opposes renal carcinoma progression. Nature. 2014;513:251–5.CrossRefPubMedPubMedCentral
16.
go back to reference Shi S, Ji S, Qin Y, Xu J, Zhang B, Xu W, Liu J, Long J, Liu C, Liu L, et al. Metabolic tumor burden is associated with major oncogenomic alterations and serum tumor markers in patients with resected pancreatic cancer. Cancer Lett. 2015;360:227–33.CrossRefPubMed Shi S, Ji S, Qin Y, Xu J, Zhang B, Xu W, Liu J, Long J, Liu C, Liu L, et al. Metabolic tumor burden is associated with major oncogenomic alterations and serum tumor markers in patients with resected pancreatic cancer. Cancer Lett. 2015;360:227–33.CrossRefPubMed
17.
go back to reference Jin X, Pan Y, Wang L, Ma T, Zhang L, Tang AH, Billadeau DD, Wu H, Huang H. Fructose-1,6-bisphosphatase inhibits ERK activation and bypasses gemcitabine resistance in pancreatic Cancer by blocking IQGAP1-MAPK interaction. Cancer Res. 2017;77:4328–41.CrossRefPubMedPubMedCentral Jin X, Pan Y, Wang L, Ma T, Zhang L, Tang AH, Billadeau DD, Wu H, Huang H. Fructose-1,6-bisphosphatase inhibits ERK activation and bypasses gemcitabine resistance in pancreatic Cancer by blocking IQGAP1-MAPK interaction. Cancer Res. 2017;77:4328–41.CrossRefPubMedPubMedCentral
18.
go back to reference Jin X, Yang C, Fan P, Xiao J, Zhang W, Zhan S, Liu T, Wang D, Wu H. CDK5/FBW7-dependent ubiquitination and degradation of EZH2 inhibits pancreatic cancer cell migration and invasion. J Biol Chem. 2017;292:6269–80.CrossRefPubMedPubMedCentral Jin X, Yang C, Fan P, Xiao J, Zhang W, Zhan S, Liu T, Wang D, Wu H. CDK5/FBW7-dependent ubiquitination and degradation of EZH2 inhibits pancreatic cancer cell migration and invasion. J Biol Chem. 2017;292:6269–80.CrossRefPubMedPubMedCentral
19.
go back to reference Jin X, Yan Y, Wang D, Ding D, Ma T, Ye Z, Jimenez R, Wang L, Wu H, Huang H. DUB3 promotes BET inhibitor resistance and Cancer progression by deubiquitinating BRD4. Mol Cell. 2018; Jin X, Yan Y, Wang D, Ding D, Ma T, Ye Z, Jimenez R, Wang L, Wu H, Huang H. DUB3 promotes BET inhibitor resistance and Cancer progression by deubiquitinating BRD4. Mol Cell. 2018;
20.
go back to reference Yang J, Jin X, Yan Y, Shao Y, Pan Y, Roberts LR, Zhang J, Huang H, Jiang J. Inhibiting histone deacetylases suppresses glucose metabolism and hepatocellular carcinoma growth by restoring FBP1 expression. Sci Rep. 2017;7:43864.CrossRefPubMedPubMedCentral Yang J, Jin X, Yan Y, Shao Y, Pan Y, Roberts LR, Zhang J, Huang H, Jiang J. Inhibiting histone deacetylases suppresses glucose metabolism and hepatocellular carcinoma growth by restoring FBP1 expression. Sci Rep. 2017;7:43864.CrossRefPubMedPubMedCentral
21.
go back to reference Yu J, Li J, Chen Y, Cao W, Lu Y, Yang J, Xing E. Snail enhances glycolysis in the epithelial-mesenchymal transition process by targeting FBP1 in gastric Cancer. Cell Physiol Biochem. 2017;43:31–8.CrossRefPubMed Yu J, Li J, Chen Y, Cao W, Lu Y, Yang J, Xing E. Snail enhances glycolysis in the epithelial-mesenchymal transition process by targeting FBP1 in gastric Cancer. Cell Physiol Biochem. 2017;43:31–8.CrossRefPubMed
22.
go back to reference Dong C, Yuan T, Wu Y, Wang Y, Fan TW, Miriyala S, Lin Y, Yao J, Shi J, Kang T, et al. Loss of FBP1 by snail-mediated repression provides metabolic advantages in basal-like breast cancer. Cancer Cell. 2013;23:316–31.CrossRefPubMedPubMedCentral Dong C, Yuan T, Wu Y, Wang Y, Fan TW, Miriyala S, Lin Y, Yao J, Shi J, Kang T, et al. Loss of FBP1 by snail-mediated repression provides metabolic advantages in basal-like breast cancer. Cancer Cell. 2013;23:316–31.CrossRefPubMedPubMedCentral
23.
go back to reference Filippakopoulos P, Qi J, Picaud S, Shen Y, Smith WB, Fedorov O, Morse EM, Keates T, Hickman TT, Felletar I, et al. Selective inhibition of BET bromodomains. Nature. 2010;468:1067–73.CrossRefPubMedPubMedCentral Filippakopoulos P, Qi J, Picaud S, Shen Y, Smith WB, Fedorov O, Morse EM, Keates T, Hickman TT, Felletar I, et al. Selective inhibition of BET bromodomains. Nature. 2010;468:1067–73.CrossRefPubMedPubMedCentral
24.
go back to reference Zhang P, Wang D, Zhao Y, Ren S, Gao K, Ye Z, Wang S, Pan CW, Zhu Y, Yan Y, et al. Intrinsic BET inhibitor resistance in SPOP-mutated prostate cancer is mediated by BET protein stabilization and AKT-mTORC1 activation. Nat Med. 2017;23:1055–62.CrossRefPubMedPubMedCentral Zhang P, Wang D, Zhao Y, Ren S, Gao K, Ye Z, Wang S, Pan CW, Zhu Y, Yan Y, et al. Intrinsic BET inhibitor resistance in SPOP-mutated prostate cancer is mediated by BET protein stabilization and AKT-mTORC1 activation. Nat Med. 2017;23:1055–62.CrossRefPubMedPubMedCentral
25.
go back to reference Yan Y, An J, Yang Y, Wu D, Bai Y, Cao W, Ma L, Chen J, Yu Z, He Y, et al. Dual inhibition of AKT-mTOR and AR signaling by targeting HDAC3 in PTEN- or SPOP-mutated prostate cancer. EMBO Mol Med. 2018;10 Yan Y, An J, Yang Y, Wu D, Bai Y, Cao W, Ma L, Chen J, Yu Z, He Y, et al. Dual inhibition of AKT-mTOR and AR signaling by targeting HDAC3 in PTEN- or SPOP-mutated prostate cancer. EMBO Mol Med. 2018;10
26.
go back to reference Kimura Y, Tanaka K. Regulatory mechanisms involved in the control of ubiquitin homeostasis. J Biochem. 2010;147:793–8.CrossRefPubMed Kimura Y, Tanaka K. Regulatory mechanisms involved in the control of ubiquitin homeostasis. J Biochem. 2010;147:793–8.CrossRefPubMed
27.
go back to reference Jin X, Pan Y, Wang L, Zhang L, Ravichandran R, Potts PR, Jiang J, Wu H, Huang H. MAGE-TRIM28 complex promotes the Warburg effect and hepatocellular carcinoma progression by targeting FBP1 for degradation. Oncogenesis. 2017;6:e312.CrossRefPubMedPubMedCentral Jin X, Pan Y, Wang L, Zhang L, Ravichandran R, Potts PR, Jiang J, Wu H, Huang H. MAGE-TRIM28 complex promotes the Warburg effect and hepatocellular carcinoma progression by targeting FBP1 for degradation. Oncogenesis. 2017;6:e312.CrossRefPubMedPubMedCentral
28.
go back to reference Bos JL. Ras oncogenes in human cancer: a review. Cancer Res. 1989;49:4682–9.PubMed Bos JL. Ras oncogenes in human cancer: a review. Cancer Res. 1989;49:4682–9.PubMed
29.
go back to reference Tsai WB, Aiba I, Long Y, Lin HK, Feun L, Savaraj N, Kuo MT. Activation of Ras/PI3K/ERK pathway induces c-Myc stabilization to upregulate argininosuccinate synthetase, leading to arginine deiminase resistance in melanoma cells. Cancer Res. 2012;72:2622–33.CrossRefPubMedPubMedCentral Tsai WB, Aiba I, Long Y, Lin HK, Feun L, Savaraj N, Kuo MT. Activation of Ras/PI3K/ERK pathway induces c-Myc stabilization to upregulate argininosuccinate synthetase, leading to arginine deiminase resistance in melanoma cells. Cancer Res. 2012;72:2622–33.CrossRefPubMedPubMedCentral
30.
go back to reference Hayes TK, Neel NF, Hu C, Gautam P, Chenard M, Long B, Aziz M, Kassner M, Bryant KL, Pierobon M, et al. Long-term ERK inhibition in KRAS-mutant pancreatic Cancer is associated with MYC degradation and senescence-like growth suppression. Cancer Cell. 2016;29:75–89.CrossRefPubMed Hayes TK, Neel NF, Hu C, Gautam P, Chenard M, Long B, Aziz M, Kassner M, Bryant KL, Pierobon M, et al. Long-term ERK inhibition in KRAS-mutant pancreatic Cancer is associated with MYC degradation and senescence-like growth suppression. Cancer Cell. 2016;29:75–89.CrossRefPubMed
31.
go back to reference Togel L, Nightingale R, Chueh AC, Jayachandran A, Tran H, Phesse T, Wu R, Sieber OM, Arango D, Dhillon AS, et al. Dual targeting of Bromodomain and Extraterminal domain proteins, and WNT or MAPK signaling, inhibits c-MYC expression and proliferation of colorectal Cancer cells. Mol Cancer Ther. 2016;15:1217–26.CrossRefPubMed Togel L, Nightingale R, Chueh AC, Jayachandran A, Tran H, Phesse T, Wu R, Sieber OM, Arango D, Dhillon AS, et al. Dual targeting of Bromodomain and Extraterminal domain proteins, and WNT or MAPK signaling, inhibits c-MYC expression and proliferation of colorectal Cancer cells. Mol Cancer Ther. 2016;15:1217–26.CrossRefPubMed
32.
go back to reference Zeller KI, Jegga AG, Aronow BJ, O'Donnell KA, Dang CV. An integrated database of genes responsive to the Myc oncogenic transcription factor: identification of direct genomic targets. Genome Biol. 2003;4:R69.CrossRefPubMedPubMedCentral Zeller KI, Jegga AG, Aronow BJ, O'Donnell KA, Dang CV. An integrated database of genes responsive to the Myc oncogenic transcription factor: identification of direct genomic targets. Genome Biol. 2003;4:R69.CrossRefPubMedPubMedCentral
33.
go back to reference Li J, Wang Y, Li QG, Xue JJ, Wang Z, Yuan X, Tong JD, Xu LC. Downregulation of FBP1 promotes tumor metastasis and indicates poor prognosis in gastric Cancer via regulating epithelial-mesenchymal transition. PLoS One. 2016;11:e0167857.CrossRefPubMedPubMedCentral Li J, Wang Y, Li QG, Xue JJ, Wang Z, Yuan X, Tong JD, Xu LC. Downregulation of FBP1 promotes tumor metastasis and indicates poor prognosis in gastric Cancer via regulating epithelial-mesenchymal transition. PLoS One. 2016;11:e0167857.CrossRefPubMedPubMedCentral
34.
go back to reference Cong J, Wang X, Zheng X, Wang D, Fu B, Sun R, Tian Z, Wei H. Dysfunction of natural killer cells by FBP1-induced inhibition of glycolysis during lung Cancer progression. Cell Metab. 2018;28:243–55. e245CrossRefPubMed Cong J, Wang X, Zheng X, Wang D, Fu B, Sun R, Tian Z, Wei H. Dysfunction of natural killer cells by FBP1-induced inhibition of glycolysis during lung Cancer progression. Cell Metab. 2018;28:243–55. e245CrossRefPubMed
37.
go back to reference Fong CY, Gilan O, Lam EY, Rubin AF, Ftouni S, Tyler D, Stanley K, Sinha D, Yeh P, Morison J, et al. BET inhibitor resistance emerges from leukaemia stem cells. Nature. 2015;525:538–42.CrossRefPubMedPubMedCentral Fong CY, Gilan O, Lam EY, Rubin AF, Ftouni S, Tyler D, Stanley K, Sinha D, Yeh P, Morison J, et al. BET inhibitor resistance emerges from leukaemia stem cells. Nature. 2015;525:538–42.CrossRefPubMedPubMedCentral
38.
go back to reference Shu S, Lin CY, He HH, Witwicki RM, Tabassum DP, Roberts JM, Janiszewska M, Huh SJ, Liang Y, Ryan J, et al. Response and resistance to BET bromodomain inhibitors in triple-negative breast cancer. Nature. 2016;529:413–7.CrossRefPubMedPubMedCentral Shu S, Lin CY, He HH, Witwicki RM, Tabassum DP, Roberts JM, Janiszewska M, Huh SJ, Liang Y, Ryan J, et al. Response and resistance to BET bromodomain inhibitors in triple-negative breast cancer. Nature. 2016;529:413–7.CrossRefPubMedPubMedCentral
39.
go back to reference Janouskova H, El Tekle G, Bellini E, Udeshi ND, Rinaldi A, Ulbricht A, Bernasocchi T, Civenni G, Losa M, Svinkina T, et al. Opposing effects of cancer-type-specific SPOP mutants on BET protein degradation and sensitivity to BET inhibitors. Nat Med. 2017;23:1046–54.CrossRefPubMedPubMedCentral Janouskova H, El Tekle G, Bellini E, Udeshi ND, Rinaldi A, Ulbricht A, Bernasocchi T, Civenni G, Losa M, Svinkina T, et al. Opposing effects of cancer-type-specific SPOP mutants on BET protein degradation and sensitivity to BET inhibitors. Nat Med. 2017;23:1046–54.CrossRefPubMedPubMedCentral
40.
go back to reference Dai X, Gan W, Li X, Wang S, Zhang W, Huang L, Liu S, Zhong Q, Guo J, Zhang J, et al. Prostate cancer-associated SPOP mutations confer resistance to BET inhibitors through stabilization of BRD4. Nat Med. 2017;23:1063–71.CrossRefPubMedPubMedCentral Dai X, Gan W, Li X, Wang S, Zhang W, Huang L, Liu S, Zhong Q, Guo J, Zhang J, et al. Prostate cancer-associated SPOP mutations confer resistance to BET inhibitors through stabilization of BRD4. Nat Med. 2017;23:1063–71.CrossRefPubMedPubMedCentral
41.
go back to reference Bandukwala HS, Gagnon J, Togher S, Greenbaum JA, Lamperti ED, Parr NJ, Molesworth AM, Smithers N, Lee K, Witherington J, et al. Selective inhibition of CD4+ T-cell cytokine production and autoimmunity by BET protein and c-Myc inhibitors. Proc Natl Acad Sci U S A. 2012;109:14532–7.CrossRefPubMedPubMedCentral Bandukwala HS, Gagnon J, Togher S, Greenbaum JA, Lamperti ED, Parr NJ, Molesworth AM, Smithers N, Lee K, Witherington J, et al. Selective inhibition of CD4+ T-cell cytokine production and autoimmunity by BET protein and c-Myc inhibitors. Proc Natl Acad Sci U S A. 2012;109:14532–7.CrossRefPubMedPubMedCentral
42.
go back to reference Ott CJ, Kopp N, Bird L, Paranal RM, Qi J, Bowman T, Rodig SJ, Kung AL, Bradner JE, Weinstock DM. BET bromodomain inhibition targets both c-Myc and IL7R in high-risk acute lymphoblastic leukemia. Blood. 2012;120:2843–52.CrossRefPubMedPubMedCentral Ott CJ, Kopp N, Bird L, Paranal RM, Qi J, Bowman T, Rodig SJ, Kung AL, Bradner JE, Weinstock DM. BET bromodomain inhibition targets both c-Myc and IL7R in high-risk acute lymphoblastic leukemia. Blood. 2012;120:2843–52.CrossRefPubMedPubMedCentral
Metadata
Title
FBP1 loss contributes to BET inhibitors resistance by undermining c-Myc expression in pancreatic ductal adenocarcinoma
Authors
Bo Wang
Ping Fan
Jingyuan Zhao
Heyu Wu
Xin Jin
Heshui Wu
Publication date
01-12-2018
Publisher
BioMed Central
Published in
Journal of Experimental & Clinical Cancer Research / Issue 1/2018
Electronic ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-018-0888-y

Other articles of this Issue 1/2018

Journal of Experimental & Clinical Cancer Research 1/2018 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine