Pooled Genomic Screens Identify Anti-apoptotic Genes as Targetable Mediators of Chemotherapy Resistance in Ovarian Cancer

High-grade serous ovarian cancer (HGSOC) is often sensitive to initial treatment with platinum and taxane combination chemotherapy, but most patients relapse with chemotherapy-resistant disease. To systematically identify genes modulating chemotherapy response, we performed pooled functional genomic screens in HGSOC cell lines treated with cisplatin, paclitaxel, or cisplatin plus paclitaxel. Genes in the intrinsic pathway of apoptosis were among the top candidate resistance genes in both gain-of-function and loss-of-function screens. In an open reading frame overexpression screen, followed by a mini-pool secondary screen, anti-apoptotic genes including BCL2L1 (BCL-XL) and BCL2L2 (BCL-W) were associated with chemotherapy resistance. In a CRISPR-Cas9 knockout screen, loss of BCL2L1 decreased cell survival whereas loss of proapoptotic genes promoted resistance. To dissect the role of individual anti-apoptotic proteins in HGSOC chemotherapy response, we evaluated overexpression or inhibition of BCL-2, BCL-XL, BCL-W, and MCL1 in HGSOC cell lines. Overexpression of anti-apoptotic proteins decreased apoptosis and modestly increased cell viability upon cisplatin or paclitaxel treatment. Conversely, specific inhibitors of BCL-XL, MCL1, or BCL-XL/BCL-2, but not BCL-2 alone, enhanced cell death when combined with cisplatin or paclitaxel. Anti-apoptotic protein inhibitors also sensitized HGSOC cells to the poly (ADP-ribose) polymerase inhibitor olaparib. These unbiased screens highlight anti-apoptotic proteins as mediators of chemotherapy resistance in HGSOC, and support inhibition of BCL-XL and MCL1, alone or combined with chemotherapy or targeted agents, in treatment of primary and recurrent HGSOC. Implications: Anti-apoptotic proteins modulate drug resistance in ovarian cancer, and inhibitors of BCL-XL or MCL1 promote cell death in combination with chemotherapy.

[1]  G. Mills,et al.  Combined MEK and BCL-2/XL Inhibition Is Effective in High-Grade Serous Ovarian Cancer Patient–Derived Xenograft Models and BIM Levels Are Predictive of Responsiveness , 2019, Molecular Cancer Therapeutics.

[2]  D. Root,et al.  DYNLL1 binds to MRE11 to limit DNA end resection in BRCA1-deficient cells , 2018, Nature.

[3]  Ashton C. Berger,et al.  A Comprehensive Pan-Cancer Molecular Study of Gynecologic and Breast Cancers. , 2018, Cancer cell.

[4]  E. Swisher,et al.  Multifaceted Impact of MicroRNA 493-5p on Genome-Stabilizing Pathways Induces Platinum and PARP Inhibitor Resistance in BRCA2-Mutated Carcinomas. , 2018, Cell reports.

[5]  Zhenfeng Zhang,et al.  Deubiquitinase USP13 dictates MCL1 stability and sensitivity to BH3 mimetic inhibitors , 2018, Nature Communications.

[6]  D. Root,et al.  Pooled Lentiviral‐Delivery Genetic Screens , 2018, Current protocols in molecular biology.

[7]  A. Leary,et al.  973PA GINECO phase II study of Navitoclax (ABT 263) in women with platinum resistant/refractory recurrent ovarian cancer (ROC) , 2017 .

[8]  G. Mills,et al.  Systems analysis of apoptotic priming in ovarian cancer identifies vulnerabilities and predictors of drug response , 2017, Nature Communications.

[9]  E. Kohn,et al.  Apoptosis is augmented in high-grade serous ovarian cancer by the combined inhibition of Bcl-2/Bcl-xL and PARP , 2017, International journal of oncology.

[10]  A. Ashkenazi,et al.  From basic apoptosis discoveries to advanced selective BCL-2 family inhibitors , 2017, Nature Reviews Drug Discovery.

[11]  A. Strasser,et al.  The MCL1 inhibitor S63845 is tolerable and effective in diverse cancer models , 2016, Nature.

[12]  D. Etemadmoghadam,et al.  Selective Targeting of Cyclin E1-Amplified High-Grade Serous Ovarian Cancer by Cyclin-Dependent Kinase 2 and AKT Inhibition , 2016, Clinical Cancer Research.

[13]  R. Tang,et al.  BCL-W is a regulator of microtubule inhibitor-induced mitotic cell death , 2016, Oncotarget.

[14]  A. Richardson,et al.  Antagonism of Bcl-XL is necessary for synergy between carboplatin and BH3 mimetics in ovarian cancer cells , 2016, Journal of Ovarian Research.

[15]  Meagan E. Sullender,et al.  Optimized sgRNA design to maximize activity and minimize off-target effects of CRISPR-Cas9 , 2015, Nature Biotechnology.

[16]  Joshua A. Bittker,et al.  Correlating chemical sensitivity and basal gene expression reveals mechanism of action , 2015, Nature chemical biology.

[17]  L. Wessels,et al.  Subunit composition of VRAC channels determines substrate specificity and cellular resistance to Pt‐based anti‐cancer drugs , 2015, The EMBO journal.

[18]  G. Shapiro,et al.  Homologous Recombination Deficiency: Exploiting the Fundamental Vulnerability of Ovarian Cancer. , 2015, Cancer discovery.

[19]  C. Ricciardelli,et al.  The role of ABC transporters in ovarian cancer progression and chemoresistance. , 2015, Critical reviews in oncology/hematology.

[20]  Joshy George,et al.  Whole–genome characterization of chemoresistant ovarian cancer , 2015, Nature.

[21]  Dolores Diaz,et al.  Exploiting selective BCL-2 family inhibitors to dissect cell survival dependencies and define improved strategies for cancer therapy , 2015, Science Translational Medicine.

[22]  S. Lheureux,et al.  Identification of predictive factors of response to the BH3‐mimetic molecule ABT‐737: An ex vivo experiment in human serous ovarian carcinoma , 2015, International journal of cancer.

[23]  Michael R. Green,et al.  Resistance to therapy in BRCA2 mutant cells due to loss of the nucleosome remodeling factor CHD4 , 2015, Genes & development.

[24]  Karen Cichowski,et al.  Drug-Induced Death Signaling Strategy Rapidly Predicts Cancer Response to Chemotherapy , 2015, Cell.

[25]  Xian-Jin Xie,et al.  Genome‐wide siRNA screen reveals coupling between mitotic apoptosis and adaptation , 2014, The EMBO journal.

[26]  S. Lheureux,et al.  PI3K/mTOR dual inhibitor NVP-BEZ235 decreases Mcl-1 expression and sensitizes ovarian carcinoma cells to Bcl-xL-targeting strategies, provided that Bim expression is induced. , 2014, Cancer letters.

[27]  Mark E. Burkard,et al.  Cytotoxicity of Paclitaxel in Breast Cancer Is due to Chromosome Missegregation on Multipolar Spindles , 2014, Science Translational Medicine.

[28]  A. Letai,et al.  Mitochondria: gatekeepers of response to chemotherapy. , 2013, Trends in cell biology.

[29]  C. Sander,et al.  Evaluating cell lines as tumour models by comparison of genomic profiles , 2013, Nature Communications.

[30]  A. Richardson,et al.  Navitoclax augments the activity of carboplatin and paclitaxel combinations in ovarian cancer cells. , 2013, Gynecologic oncology.

[31]  S. Lheureux,et al.  Platinum compounds sensitize ovarian carcinoma cells to ABT-737 by modulation of the Mcl-1/Noxa axis , 2013, Apoptosis.

[32]  Andrew L. Kung,et al.  Chemical genomics identifies small-molecule MCL1 repressors and BCL-xL as a predictor of MCL1 dependency. , 2012, Cancer cell.

[33]  Adam A. Margolin,et al.  The Cancer Cell Line Encyclopedia enables predictive modeling of anticancer drug sensitivity , 2012, Nature.

[34]  F. Peale,et al.  Navitoclax (ABT-263) Reduces Bcl-xL–Mediated Chemoresistance in Ovarian Cancer Models , 2012, Molecular Cancer Therapeutics.

[35]  A. Letai,et al.  Pretreatment Mitochondrial Priming Correlates with Clinical Response to Cytotoxic Chemotherapy , 2011, Science.

[36]  R. Bast,et al.  Modulating microtubule stability enhances the cytotoxic response of cancer cells to Paclitaxel. , 2011, Cancer research.

[37]  Rochelle L. Garcia,et al.  Secondary somatic mutations restoring BRCA1/2 predict chemotherapy resistance in hereditary ovarian carcinomas. , 2011, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[38]  Thomas M Green,et al.  A public genome-scale lentiviral expression library of human ORFs , 2011, Nature Methods.

[39]  Adam R. Johnson,et al.  Sensitivity to antitubulin chemotherapeutics is regulated by MCL1 and FBW7 , 2011, Nature.

[40]  Shilpi Arora,et al.  RNAi screening of the kinome identifies modulators of cisplatin response in ovarian cancer cells. , 2010, Gynecologic oncology.

[41]  Derek Y. Chiang,et al.  The landscape of somatic copy-number alteration across human cancers , 2010, Nature.

[42]  L. Poulain,et al.  Bcl‐XL and MCL‐1 constitute pertinent targets in ovarian carcinoma and their concomitant inhibition is sufficient to induce apoptosis , 2009, International journal of cancer.

[43]  W. Wilson,et al.  Ongoing phase I studies of ABT-263: Mitigating Bcl-XL induced thrombocytopenia with lead-in and continuous dosing. , 2009, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[44]  S. Kaye,et al.  The Bcl-2/Bcl-XL Family Inhibitor ABT-737 Sensitizes Ovarian Cancer Cells to Carboplatin , 2007, Clinical Cancer Research.

[45]  A. Piché,et al.  Bcl-XL is functionally non-equivalent for the regulation of growth and survival in human ovarian cancer cells , 2006 .

[46]  K. Griffith,et al.  Expression of Bcl-xL in ovarian carcinoma is associated with chemoresistance and recurrent disease. , 2005, Gynecologic oncology.

[47]  O. Katoh,et al.  Increased MCL–1 Expression Is Associated with Poor Prognosis in Ovarian Carcinomas , 2002, Japanese journal of cancer research : Gann.

[48]  I. Nagata,et al.  Bcl-2 as a predictor of chemosensitivity and prognosis in primary epithelial ovarian cancer. , 1999, European journal of cancer.

[49]  S. Kassim,et al.  Increased bcl-2 expression is associated with primary resistance to chemotherapy in human epithelial ovarian cancer. , 1999, Clinical biochemistry.

[50]  C. Hu,et al.  Bcl-xL is expressed in ovarian carcinoma and modulates chemotherapy-induced apoptosis. , 1998, Gynecologic oncology.

[51]  A. Piché,et al.  Bcl-X(L) is functionally non-equivalent for the regulation of growth and survival in human ovarian cancer cells. , 2006, Gynecologic oncology.