Systematic identification of genomic markers of drug sensitivity in cancer cells

Mathew J Garnett, Elena J Edelman, Sonja J Heidorn, Chris D Greenman, Anahita Dastur, King Wai Lau, Patricia Greninger, I Richard Thompson, Xi Luo, Jorge Soares, Qingsong Liu, Francesco Iorio, Didier Surdez, Li Chen, Randy J Milano, Graham R Bignell, Ah T Tam, Helen Davies, Jesse A Stevenson, Syd Barthorpe, Stephen R Lutz, Fiona Kogera, Karl Lawrence, Anne McLaren-Douglas, Xeni Mitropoulos, Tatiana Mironenko, Helen Thi, Laura Richardson, Wenjun Zhou, Frances Jewitt, Tinghu Zhang, Patrick O'Brien, Jessica L Boisvert, Stacey Price, Wooyoung Hur, Wanjuan Yang, Xianming Deng, Adam Butler, Hwan Geun Choi, Jae Won Chang, Jose Baselga, Ivan Stamenkovic, Jeffrey A Engelman, Sreenath V Sharma, Olivier Delattre, Julio Saez-Rodriguez, Nathanael S Gray, Jeffrey Settleman, P Andrew Futreal, Daniel A Haber, Michael R Stratton, Sridhar Ramaswamy, Ultan McDermott, Cyril H Benes, Mathew J Garnett, Elena J Edelman, Sonja J Heidorn, Chris D Greenman, Anahita Dastur, King Wai Lau, Patricia Greninger, I Richard Thompson, Xi Luo, Jorge Soares, Qingsong Liu, Francesco Iorio, Didier Surdez, Li Chen, Randy J Milano, Graham R Bignell, Ah T Tam, Helen Davies, Jesse A Stevenson, Syd Barthorpe, Stephen R Lutz, Fiona Kogera, Karl Lawrence, Anne McLaren-Douglas, Xeni Mitropoulos, Tatiana Mironenko, Helen Thi, Laura Richardson, Wenjun Zhou, Frances Jewitt, Tinghu Zhang, Patrick O'Brien, Jessica L Boisvert, Stacey Price, Wooyoung Hur, Wanjuan Yang, Xianming Deng, Adam Butler, Hwan Geun Choi, Jae Won Chang, Jose Baselga, Ivan Stamenkovic, Jeffrey A Engelman, Sreenath V Sharma, Olivier Delattre, Julio Saez-Rodriguez, Nathanael S Gray, Jeffrey Settleman, P Andrew Futreal, Daniel A Haber, Michael R Stratton, Sridhar Ramaswamy, Ultan McDermott, Cyril H Benes

Abstract

Clinical responses to anticancer therapies are often restricted to a subset of patients. In some cases, mutated cancer genes are potent biomarkers for responses to targeted agents. Here, to uncover new biomarkers of sensitivity and resistance to cancer therapeutics, we screened a panel of several hundred cancer cell lines--which represent much of the tissue-type and genetic diversity of human cancers--with 130 drugs under clinical and preclinical investigation. In aggregate, we found that mutated cancer genes were associated with cellular response to most currently available cancer drugs. Classic oncogene addiction paradigms were modified by additional tissue-specific or expression biomarkers, and some frequently mutated genes were associated with sensitivity to a broad range of therapeutic agents. Unexpected relationships were revealed, including the marked sensitivity of Ewing's sarcoma cells harbouring the EWS (also known as EWSR1)-FLI1 gene translocation to poly(ADP-ribose) polymerase (PARP) inhibitors. By linking drug activity to the functional complexity of cancer genomes, systematic pharmacogenomic profiling in cancer cell lines provides a powerful biomarker discovery platform to guide rational cancer therapeutic strategies.

Figures

Figure 1. A systematic screen in cancer…
Figure 1. A systematic screen in cancer cell lines identifies therapeutic biomarkers
a, The number of tumour-derived cell lines used for screening classified according to tissue type (n = 639 in total). b, The panel of 130 screening drugs classified according to their therapeutic targets, primary effector pathways, and cellular functions. A single drug may be included in multiple categories. The inset indicates the number of drugs screened against a selection of prototype cancer targets. c, A volcano plot representation of MANOVA results showing the magnitude (effect; x-axis) and significance (p-value; inverted y-axis) of all drug-gene associations. Each circle represents a single drug-gene interaction and the size is proportional to the number of mutant cell lines screened (range 1 – 334). The horizontal dashed line indicates the threshold of statistical significance (0.2 FDR, P < 0.0099). Insets I and II are magnified views of selected highly significant associations and the drug name, therapeutically relevant target(s) (in superscript), and cancer gene (in brackets) are given for each. The p-values for nilotinibABL(BCR-ABL), P = 2.54 × 10−65, and nutlin-3aMDM2(TP53), P= 2.78 × 10−37, have been capped at 1 × 10−28 in this representation.
Figure 2. Biomarkers of drug sensitivity and…
Figure 2. Biomarkers of drug sensitivity and resistance
a, Gene-specific volcano plots of drug sensitivity associated with BRAF mutations in cancer cell lines (n = 54). b-k, Scatter plots of cell line IC50 (uM) values from selected drug-gene associations. IC50 values are on a log scale comparing mutated or non-mutated (WT) cell lines. Each circle represents the IC50 of one cell line and the red bar is the geometric mean. The drug name is indicated above each plot and therapeutic drug target(s) are bracketed.
Figure 3. Multi-feature genomic signatures of drug…
Figure 3. Multi-feature genomic signatures of drug response
a, The top drug-feature associations identified by the EN are plotted for their frequency and effect size. Associations are colored black for expression features, red for mutations, blue for copy number, and green for tissue. b-c. Heatmaps of highly significant EN features associated with response to b, dasatinib (inhibitor of SRC,ABL) and c, 17-AAG (HSP90 inhibitor) for the 14 most sensitive (purple) and resistant (yellow) cell lines. For each cell line mutation features are at the top of the heatmap shown in black (present) or gray (absent), followed by expression features (blue corresponds to lower expression, red to higher expression). To the left of each feature is a bar indicating the absolute value of the effect size. Bars in purple are negative effects, indicating features associated with sensitivity, and bars in yellow are positive effects, indicating features associated with resistance. The natural log IC50 values are represented at the bottom. For clarity, only the top 4 features associated with sensitivity and resistance to 17-AAG are shown.
Figure 4. Ewing’s sarcoma cell lines are…
Figure 4. Ewing’s sarcoma cell lines are sensitive to PARP inhibition
a, The IC50 values of WT and EWS-FLI1 fusion positive cell lines to olaparib and AG-014699. b, Dose response curves to olaparib following 6-days constant drug exposure. Cell lines are classified according to tissue sub-type. c, Colony formation assays were performed for 7-21 days over a range of olaparib concentrations (0.1, 0.32, 1, 3.2 or 10 uM) and the concentration at which the number of colonies is reduced >90% for each cell line is indicated. d, Olaparib induced apoptosis in Ewing’s sarcoma cell lines following 72 hours treatment. e, Sensitivity to olaparib of EWS-FLI1 and FUS-CHOP transformed mouse mesenchymal cells compared to the SK-N-MC cell line (which harbors the EWS-FLI1 fusion). f, Sensitivity to olaparib of A673 cells transiently transfected with (siEF1) and without (siCT) EWS-FLI1 specific siRNA. All error bars are s.d from triplicate measurements except for b where error bars have been removed for clarity.

References

    1. Druker BJ, et al. Five-year follow-up of patients receiving imatinib for chronic myeloid leukemia. N Engl J Med. 2006;355:2408–2417.
    1. Kwak EL, et al. Anaplastic lymphoma kinase inhibition in non-small-cell lung cancer. N Engl J Med. 2010;363:1693–1703.
    1. Chapman PB, et al. Improved Survival with Vemurafenib in Melanoma with BRAF V600E Mutation. N Engl J Med. 2011
    1. McDermott U, Settleman J. Personalized cancer therapy with selective kinase inhibitors: an emerging paradigm in medical oncology. J Clin Oncol. 2009;27:5650–5659.
    1. Shoemaker RH, et al. Development of human tumor cell line panels for use in disease-oriented drug screening. Prog Clin Biol Res. 1988;276:265–286.
    1. Weinstein JN, et al. An information-intensive approach to the molecular pharmacology of cancer. Science. 1997;275:343–349.
    1. McDermott U, et al. Identification of genotype-correlated sensitivity to selective kinase inhibitors by using high-throughput tumor cell line profiling. Proc Natl Acad Sci U S A. 2007;104:19936–19941.
    1. Suwaki N, et al. A HIF-regulated VHL-PTP1B-Src signaling axis identifies a therapeutic target in renal cell carcinoma. Sci Transl Med. 2011;3:85ra47. doi:10.1126/scitranslmed.3002004.
    1. Deng L, et al. Rho-kinase inhibitor, fasudil, suppresses glioblastoma cell line progression in vitro and in vivo. Cancer Biol Ther. 2010;9:875–884.
    1. Weber DM, et al. Lenalidomide plus dexamethasone for relapsed multiple myeloma in North America. N Engl J Med. 2007;357:2133–2142. doi:10.1056/NEJMoa070596.
    1. Konecny GE, et al. Activity of the dual kinase inhibitor lapatinib (GW572016) against HER-2-overexpressing and trastuzumab-treated breast cancer cells. Cancer Res. 2006;66:1630–1639.
    1. Lynch TJ, et al. Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N Engl J Med. 2004;350:2129–2139.
    1. O’Farrell AM, et al. SU11248 is a novel FLT3 tyrosine kinase inhibitor with potent activity in vitro and in vivo. Blood. 2003;101:3597–3605.
    1. Kunii K, et al. FGFR2-amplified gastric cancer cell lines require FGFR2 and Erbb3 signaling for growth and survival. Cancer Res. 2008;68:2340–2348.
    1. Byron SA, et al. Inhibition of activated fibroblast growth factor receptor 2 in endometrial cancer cells induces cell death despite PTEN abrogation. Cancer Res. 2008;68:6902–6907. doi:10.1158/0008-5472.CAN-08-0770.
    1. Vassilev LT, et al. In vivo activation of the p53 pathway by small-molecule antagonists of MDM2. Science. 2004;303:844–848. doi:10.1126/science.1092472.
    1. Konecny GE, et al. Expression of p16 and retinoblastoma determines response to CDK4/6 inhibition in ovarian cancer. Clinical cancer research: an official journal of the American Association for Cancer Research. 2011;17:1591–1602. doi:10.1158/1078-0432.CCR-10-2307.
    1. Boisvert-Adamo K, Longmate W, Abel EV, Aplin AE. Mcl-1 is required for melanoma cell resistance to anoikis. Mol Cancer Res. 2009;7:549–556.
    1. Brown CJ, Lain S, Verma CS, Fersht AR, Lane DP. Awakening guardian angels: drugging the p53 pathway. Nat Rev Cancer. 2009;9:862–873. doi:10.1038/nrc2763.
    1. Zou H, Hastie T. Regularization and variable selection via the elastic net. J.R. Statist. Soc. B. 2005;67:301–320.
    1. Hanafusa H, Torii S, Yasunaga T, Nishida E. Sprouty1 and Sprouty2 provide a control mechanism for the Ras/MAPK signalling pathway. Nature cell biology. 2002;4:850–858. doi:10.1038/ncb867.
    1. Patterson KI, Brummer T, O’Brien PM, Daly RJ. Dual-specificity phosphatases: critical regulators with diverse cellular targets. The Biochemical journal. 2009;418:475–489.
    1. Dry JR, et al. Transcriptional pathway signatures predict MEK addiction and response to selumetinib (AZD6244) Cancer Res. 2010;70:2264–2273. doi:10.1158/0008-5472.CAN-09-1577.
    1. Guo W, et al. Formation of 17-allylamino-demethoxygeldanamycin (17-AAG) hydroquinone by NAD(P)H:quinone oxidoreductase 1: role of 17-AAG hydroquinone in heat shock protein 90 inhibition. Cancer Res. 2005;65:10006–10015. doi:10.1158/0008-5472.CAN-05-2029.
    1. Fong PC, et al. Inhibition of poly(ADP-ribose) polymerase in tumors from BRCA mutation carriers. N Engl J Med. 2009;361:123–134.
    1. McCabe N, et al. BRCA2-deficient CAPAN-1 cells are extremely sensitive to the inhibition of Poly (ADP-Ribose) polymerase: an issue of potency. Cancer Biol Ther. 2005;4:934–936.
    1. Riggi N, et al. Development of Ewing’s sarcoma from primary bone marrow-derived mesenchymal progenitor cells. Cancer Res. 2005;65:11459–11468. doi:10.1158/0008-5472.CAN-05-1696.
    1. Riggi N, et al. Expression of the FUS-CHOP fusion protein in primary mesenchymal progenitor cells gives rise to a model of myxoid liposarcoma. Cancer Res. 2006;66:7016–7023. doi:10.1158/0008-5472.CAN-05-3979.
    1. Brenner JC, et al. Mechanistic Rationale for Inhibition of Poly(ADP-Ribose) Polymerase in ETS Gene Fusion-Positive Prostate Cancer. Cancer Cell. 2011;19:664–678.
    1. Balamuth NJ, Womer RB. Ewing’s sarcoma. Lancet Oncol. 2010;11:184–192.
    1. Jordi Barretina GC, et al. The Cancer Cell Line Encyclopedia: using preclinical models to predict anticancer drug sensitivity. Nature. 2012
METHODS REFERENCES
    1. Prieur A, Tirode F, Cohen P, Delattre O. EWS/FLI-1 silencing and gene profiling of Ewing cells reveal downstream oncogenic pathways and a crucial role for repression of insulin-like growth factor binding protein 3. Mol Cell Biol. 2004;24:7275–7283. doi:10.1128/MCB.24.16.7275-7283.2004.
    1. Boland CR, et al. A National Cancer Institute Workshop on Microsatellite Instability for cancer detection and familial predisposition: development of international criteria for the determination of microsatellite instability in colorectal cancer. Cancer Res. 1998;58:5248–5257.
    1. Greenman CD, et al. PICNIC: an algorithm to predict absolute allelic copy number variation with microarray cancer data. Biostatistics. 2010;11:164–175.
    1. Bolstad BM, Irizarry RA, Astrand M, Speed TP. A comparison of normalization methods for high density oligonucleotide array data based on variance and bias. Bioinformatics. 2003;19:185–193.
    1. Frey BJ, Dueck D. Clustering by passing messages between data points. Science. 2007;315:972–976. doi:10.1126/science.1136800.
    1. Iorio F, et al. Discovery of drug mode of action and drug repositioning from transcriptional responses. Proceedings of the National Academy of Sciences of the United States of America. 2010;107:14621–14626. doi:10.1073/pnas.1000138107.
    1. Shannon P, et al. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome research. 2003;13:2498–2504. doi:10.1101/gr.1239303.

Source: PubMed

3
订阅