Mutational landscape of metastatic cancer revealed from prospective clinical sequencing of 10,000 patients

Ahmet Zehir, Ryma Benayed, Ronak H Shah, Aijazuddin Syed, Sumit Middha, Hyunjae R Kim, Preethi Srinivasan, Jianjiong Gao, Debyani Chakravarty, Sean M Devlin, Matthew D Hellmann, David A Barron, Alison M Schram, Meera Hameed, Snjezana Dogan, Dara S Ross, Jaclyn F Hechtman, Deborah F DeLair, JinJuan Yao, Diana L Mandelker, Donavan T Cheng, Raghu Chandramohan, Abhinita S Mohanty, Ryan N Ptashkin, Gowtham Jayakumaran, Meera Prasad, Mustafa H Syed, Anoop Balakrishnan Rema, Zhen Y Liu, Khedoudja Nafa, Laetitia Borsu, Justyna Sadowska, Jacklyn Casanova, Ruben Bacares, Iwona J Kiecka, Anna Razumova, Julie B Son, Lisa Stewart, Tessara Baldi, Kerry A Mullaney, Hikmat Al-Ahmadie, Efsevia Vakiani, Adam A Abeshouse, Alexander V Penson, Philip Jonsson, Niedzica Camacho, Matthew T Chang, Helen H Won, Benjamin E Gross, Ritika Kundra, Zachary J Heins, Hsiao-Wei Chen, Sarah Phillips, Hongxin Zhang, Jiaojiao Wang, Angelica Ochoa, Jonathan Wills, Michael Eubank, Stacy B Thomas, Stuart M Gardos, Dalicia N Reales, Jesse Galle, Robert Durany, Roy Cambria, Wassim Abida, Andrea Cercek, Darren R Feldman, Mrinal M Gounder, A Ari Hakimi, James J Harding, Gopa Iyer, Yelena Y Janjigian, Emmet J Jordan, Ciara M Kelly, Maeve A Lowery, Luc G T Morris, Antonio M Omuro, Nitya Raj, Pedram Razavi, Alexander N Shoushtari, Neerav Shukla, Tara E Soumerai, Anna M Varghese, Rona Yaeger, Jonathan Coleman, Bernard Bochner, Gregory J Riely, Leonard B Saltz, Howard I Scher, Paul J Sabbatini, Mark E Robson, David S Klimstra, Barry S Taylor, Jose Baselga, Nikolaus Schultz, David M Hyman, Maria E Arcila, David B Solit, Marc Ladanyi, Michael F Berger, Ahmet Zehir, Ryma Benayed, Ronak H Shah, Aijazuddin Syed, Sumit Middha, Hyunjae R Kim, Preethi Srinivasan, Jianjiong Gao, Debyani Chakravarty, Sean M Devlin, Matthew D Hellmann, David A Barron, Alison M Schram, Meera Hameed, Snjezana Dogan, Dara S Ross, Jaclyn F Hechtman, Deborah F DeLair, JinJuan Yao, Diana L Mandelker, Donavan T Cheng, Raghu Chandramohan, Abhinita S Mohanty, Ryan N Ptashkin, Gowtham Jayakumaran, Meera Prasad, Mustafa H Syed, Anoop Balakrishnan Rema, Zhen Y Liu, Khedoudja Nafa, Laetitia Borsu, Justyna Sadowska, Jacklyn Casanova, Ruben Bacares, Iwona J Kiecka, Anna Razumova, Julie B Son, Lisa Stewart, Tessara Baldi, Kerry A Mullaney, Hikmat Al-Ahmadie, Efsevia Vakiani, Adam A Abeshouse, Alexander V Penson, Philip Jonsson, Niedzica Camacho, Matthew T Chang, Helen H Won, Benjamin E Gross, Ritika Kundra, Zachary J Heins, Hsiao-Wei Chen, Sarah Phillips, Hongxin Zhang, Jiaojiao Wang, Angelica Ochoa, Jonathan Wills, Michael Eubank, Stacy B Thomas, Stuart M Gardos, Dalicia N Reales, Jesse Galle, Robert Durany, Roy Cambria, Wassim Abida, Andrea Cercek, Darren R Feldman, Mrinal M Gounder, A Ari Hakimi, James J Harding, Gopa Iyer, Yelena Y Janjigian, Emmet J Jordan, Ciara M Kelly, Maeve A Lowery, Luc G T Morris, Antonio M Omuro, Nitya Raj, Pedram Razavi, Alexander N Shoushtari, Neerav Shukla, Tara E Soumerai, Anna M Varghese, Rona Yaeger, Jonathan Coleman, Bernard Bochner, Gregory J Riely, Leonard B Saltz, Howard I Scher, Paul J Sabbatini, Mark E Robson, David S Klimstra, Barry S Taylor, Jose Baselga, Nikolaus Schultz, David M Hyman, Maria E Arcila, David B Solit, Marc Ladanyi, Michael F Berger

Abstract

Tumor molecular profiling is a fundamental component of precision oncology, enabling the identification of genomic alterations in genes and pathways that can be targeted therapeutically. The existence of recurrent targetable alterations across distinct histologically defined tumor types, coupled with an expanding portfolio of molecularly targeted therapies, demands flexible and comprehensive approaches to profile clinically relevant genes across the full spectrum of cancers. We established a large-scale, prospective clinical sequencing initiative using a comprehensive assay, MSK-IMPACT, through which we have compiled tumor and matched normal sequence data from a unique cohort of more than 10,000 patients with advanced cancer and available pathological and clinical annotations. Using these data, we identified clinically relevant somatic mutations, novel noncoding alterations, and mutational signatures that were shared by common and rare tumor types. Patients were enrolled on genomically matched clinical trials at a rate of 11%. To enable discovery of novel biomarkers and deeper investigation into rare alterations and tumor types, all results are publicly accessible.

Conflict of interest statement

COMPETING FINANCIAL INTERESTS

The authors declare no competing financial interests.

Figures

Figure 1
Figure 1
Overview of MSK-IMPACT clinical workflow. Patients provide informed consent for paired tumor-normal sequence analysis, and a blood sample is collected as a source of normal DNA. DNA is extracted from tumor and blood samples using automated protocols, and sequence libraries are prepared and captured using hybridization probes targeting all coding exons of 410 genes and select introns of recurrently rearranged genes. Following sequencing, paired reads are analyzed through a custom bioinformatics pipeline that detects multiple classes of genomic rearrangements. Results are loaded into an in-house developed genomic variants database, MPath, upon which they are manually reviewed for quality and accuracy. Genomic alterations are reported in the electronic medical record, transmitted to an institutional database (Darwin) that facilitates automated clinical trial matching, and automatically uploaded to the cBioPortal for data mining and interpretation.
Figure 2
Figure 2
Overview of the MSK-IMPACT cohort. (a) Distribution of tumor types among cases successfully sequenced from 10,336 patients. Cases represented 62 principal tumor types encapsulating 361 detailed tumor types. (b) Frequency of gene alterations in TCGA and MSK-IMPACT cohorts. Genes that were statistically significantly mutated in TCGA studies are displayed, and genes that show a significant difference between the two cohorts are labeled. (c) Recurrent somatic alterations across common tumor types. Genes with a cohort-level alteration frequency of ≥5% or a tumor type-specific alteration frequency of ≥30% are displayed. Bars indicate the percent of cases within each tumor type harboring different classes of genomic alterations.
Figure 3
Figure 3
Spectrum of TERT promoter mutations in cancer. (a) Location of all TERT promoter mutations relative to the transcription start site (+1). Observed nucleotide changes leading presumptive ETS transcription binding sites are shown for the three most common mutational hotspots. Inset shows the distribution of cancer types harboring mutations at each individual hotspot. (b) Bar plot depicting the percentage of cases in each common principal tumor type (left) and melanoma sub-types (right) harboring a TERT promoter mutation. (c) Kaplan-Meier survival curves for the most prominent detailed tumor types belonging to the principal tumor types with the highest prevalence of TERT promoter mutations. Survival was measured starting from the date of the procedure to obtain the specimen sequenced. Cases where specimens were obtained more than 12 months prior to MSK-IMPACT sequencing were excluded from this analysis.
Figure 4
Figure 4
Spectrum of kinase fusions identified by MSK-IMPACT. (a) Kinase genes recurrently rearranged to form putative gene fusions including the kinase domain, displayed across principal tumor types. (b) List of fusions containing BRAF gene. * novel fusion partner; † complex fusion resolved using an orthogonal RNAseq based assay. (c) In-frame intragenic deletions observed in BRAF, encompassing 5 - 9 exons upstream of the kinase domain.
Figure 5
Figure 5
Mutational signatures derived from MSK-IMPACT targeted sequencing data. (a) Distribution of the somatic tumor mutation burden (TMB), defined as non-synonymous coding mutations per Megabase, for common principal tumor types. (b) Distribution of observed mutation rates across all tumors sequenced was used to identify a threshold of 13.8 mutations/Mb, indicative of high mutation burden. (c) Dominant mutation signatures identified in cases with high mutation burden. The percent of cases harboring a dominant mutation signature is shown for each principal tumor type. POLE: DNA Polymerase ε; MMR: Mismatch repair deficiency; UV: Ultraviolet light; TMZ: Temozolomide. (d) Individual tumors harboring dominant mutation signatures. Bar charts display the total number of coding mutations (gray) and the fraction of mutations explained by the major signature (colored). Tracks below the bar charts indicate: i) POLE mutation status, ii) MMR pathway mutation status, iii) MSIsensor score, iv) indel to SNV ratio, v) reported smoking status, and vi) cancer type. (e) Tumor type distribution for samples with a high mutation burden, dominant MMR signature, and inferred MSI. (f) 55-year-old with castrate and enzalutamide resistant prostate cancer with an MMR signature (19 mutations, including 6 frameshift indels) and no clear underlying somatic or germline MMR pathway lesion. A pathogenic germline MUTYH variant was detected, which may contribute to the MSI phenotype. Upon initiation of treatment on an anti-PD-L1 immunotherapy regimen, significant tumor regression was observed. Line charts show the relative tumor size based on RECIST criteria and serum PSA levels. MRI images show the decreasing tumor size at indicated time points (scale bar = 10 cm).
Figure 6
Figure 6
Clinical actionability of somatic alterations revealed by MSK-IMPACT. (a) Alterations were annotated based on their clinical actionability according to OncoKB, and samples were assigned the level of the most significant alteration. Briefly, levels of evidence varied according to whether mutations are FDA-recognized biomarkers (Level 1), predict response to standard-of-care therapies (Level 2), or predict for response to investigational agents in clinical trials (Level 3). Levels 2 and 3 were subdivided according to whether the evidence existed for the pertinent tumor type (2A, 3A) or a different tumor type (2B, 3B). The distribution of the highest level of actionability across all patients is displayed. (b) Distribution of levels of actionability across tumor types (GNET: gastrointestincal neuroendocrine tumor). (c) Number of patients enrolled on genomically-matched clinical trials on the basis of different gene alterations.

References

    1. Garraway LA. Genomics-driven oncology: framework for an emerging paradigm. J Clin Oncol. 2013;31:1806–14.
    1. Varghese AM, Berger MF. Advancing clinical oncology through genome biology and technology. Genome Biol. 2014;15:427.
    1. Lindeman NI, et al. Molecular testing guideline for selection of lung cancer patients for EGFR and ALK tyrosine kinase inhibitors: guideline from the College of American Pathologists, International Association for the Study of Lung Cancer, and Association for Molecular Pathology. J Mol Diagn. 2013;15:415–53.
    1. Chapman PB, et al. Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N Engl J Med. 2011;364:2507–16.
    1. Hyman DM, et al. Vemurafenib in Multiple Nonmelanoma Cancers with BRAF V600 Mutations. N Engl J Med. 2015;373:726–36.
    1. Singh RR, et al. Clinical validation of a next-generation sequencing screen for mutational hotspots in 46 cancer-related genes. J Mol Diagn. 2013;15:607–22.
    1. Roychowdhury S, et al. Personalized oncology through integrative high-throughput sequencing: a pilot study. Sci Transl Med. 2011;3:111ra121.
    1. Frampton GM, et al. Development and validation of a clinical cancer genomic profiling test based on massively parallel DNA sequencing. Nat Biotechnol. 2013;31:1023–31.
    1. Beltran H, et al. Whole-Exome Sequencing of Metastatic Cancer and Biomarkers of Treatment Response. JAMA Oncol. 2015;1:466–74.
    1. Sholl LM, et al. Institutional implementation of clinical tumor profiling on an unselected cancer population. JCI Insight. 2016;1:e87062.
    1. Cheng DT, et al. Memorial Sloan Kettering-Integrated Mutation Profiling of Actionable Cancer Targets (MSK-IMPACT): A Hybridization Capture-Based Next-Generation Sequencing Clinical Assay for Solid Tumor Molecular Oncology. J Mol Diagn. 2015;17:251–64.
    1. Cerami E, et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2012;2:401–4.
    1. Ciriello G, et al. Emerging landscape of oncogenic signatures across human cancers. Nat Genet. 2013;45:1127–33.
    1. Simen BB, et al. Validation of a next-generation-sequencing cancer panel for use in the clinical laboratory. Arch Pathol Lab Med. 2015;139:508–17.
    1. Forbes SA, et al. COSMIC: somatic cancer genetics at high-resolution. Nucleic Acids Res. 2016
    1. Kandoth C, et al. Mutational landscape and significance across 12 major cancer types. Nature. 2013;502:333–9.
    1. Cancer Genome Atlas Research N. Integrated genomic characterization of papillary thyroid carcinoma. Cell. 2014;159:676–90.
    1. Cancer Genome Atlas Research N. Comprehensive molecular characterization of gastric adenocarcinoma. Nature. 2014;513:202–9.
    1. Davis CF, et al. The somatic genomic landscape of chromophobe renal cell carcinoma. Cancer Cell. 2014;26:319–30.
    1. Powell E, Piwnica-Worms D, Piwnica-Worms H. Contribution of p53 to metastasis. Cancer Discov. 2014;4:405–14.
    1. Watson PA, Arora VK, Sawyers CL. Emerging mechanisms of resistance to androgen receptor inhibitors in prostate cancer. Nat Rev Cancer. 2015;15:701–11.
    1. Toy W, et al. ESR1 ligand-binding domain mutations in hormone-resistant breast cancer. Nat Genet. 2013;45:1439–45.
    1. Robinson DR, et al. Activating ESR1 mutations in hormone-resistant metastatic breast cancer. Nat Genet. 2013;45:1446–51.
    1. Chang MT, et al. Identifying recurrent mutations in cancer reveals widespread lineage diversity and mutational specificity. Nat Biotechnol. 2016;34:155–63.
    1. Baca SC, et al. Punctuated evolution of prostate cancer genomes. Cell. 2013;153:666–77.
    1. Horn S, et al. TERT promoter mutations in familial and sporadic melanoma. Science. 2013;339:959–61.
    1. Huang FW, et al. Highly recurrent TERT promoter mutations in human melanoma. Science. 2013;339:957–9.
    1. Killela PJ, et al. TERT promoter mutations occur frequently in gliomas and a subset of tumors derived from cells with low rates of self-renewal. Proc Natl Acad Sci U S A. 2013;110:6021–6.
    1. Gao K, et al. TERT promoter mutations and long telomere length predict poor survival and radiotherapy resistance in gliomas. Oncotarget. 2016;7:8712–25.
    1. Melo M, et al. TERT promoter mutations are a major indicator of poor outcome in differentiated thyroid carcinomas. J Clin Endocrinol Metab. 2014;99:E754–65.
    1. Piscuoglio S, et al. Massively parallel sequencing of phyllodes tumours of the breast reveals actionable mutations, and TERT promoter hotspot mutations and TERT gene amplification as likely drivers of progression. J Pathol. 2016;238:508–18.
    1. Stransky N, Cerami E, Schalm S, Kim JL, Lengauer C. The landscape of kinase fusions in cancer. Nat Commun. 2014;5:4846.
    1. Ross JS, et al. The distribution of BRAF gene fusions in solid tumors and response to targeted therapy. Int J Cancer. 2016;138:881–90.
    1. Menzies AM, et al. Clinical activity of the MEK inhibitor trametinib in metastatic melanoma containing BRAF kinase fusion. Pigment Cell Melanoma Res. 2015;28:607–10.
    1. Poulikakos PI, et al. RAF inhibitor resistance is mediated by dimerization of aberrantly spliced BRAF(V600E) Nature. 2011;480:387–90.
    1. Yao Z, et al. BRAF Mutants Evade ERK-Dependent Feedback by Different Mechanisms that Determine Their Sensitivity to Pharmacologic Inhibition. Cancer Cell. 2015;28:370–83.
    1. Alexandrov LB, et al. Signatures of mutational processes in human cancer. Nature. 2013;500:415–21.
    1. Niu B, et al. MSIsensor: microsatellite instability detection using paired tumor-normal sequence data. Bioinformatics. 2014;30:1015–6.
    1. Le DT, et al. PD-1 Blockade in Tumors with Mismatch-Repair Deficiency. N Engl J Med. 2015;372:2509–20.
    1. Chakravarty D, et al. OncoKB: A Precision Oncology Knowledge Base. J Clin Oncol Precision Oncology. 2017
    1. Meric-Bernstam F, et al. Feasibility of Large-Scale Genomic Testing to Facilitate Enrollment Onto Genomically Matched Clinical Trials. J Clin Oncol. 2015;33:2753–62.
    1. Ross JS, et al. Comprehensive Genomic Profiling of Carcinoma of Unknown Primary Site: New Routes to Targeted Therapies. JAMA Oncol. 2015;1:40–9.
    1. Zhu Z, et al. Inhibition of KRAS-driven tumorigenicity by interruption of an autocrine cytokine circuit. Cancer Discov. 2014;4:452–65.
    1. Manchado E, et al. A combinatorial strategy for treating KRAS-mutant lung cancer. Nature. 2016;534:647–51.
    1. Eubank MH, et al. Automated eligibility screening and monitoring for genotype-driven precision oncology trials. J Am Med Inform Assoc. 2016;23:777–81.
    1. Schwaederle M, et al. On the Road to Precision Cancer Medicine: Analysis of Genomic Biomarker Actionability in 439 Patients. Mol Cancer Ther. 2015;14:1488–94.
    1. Stockley TL, et al. Molecular profiling of advanced solid tumors and patient outcomes with genotype-matched clinical trials: the Princess Margaret IMPACT/COMPACT trial. Genome Med. 2016;8:109.
    1. Jones S, et al. Personalized genomic analyses for cancer mutation discovery and interpretation. Sci Transl Med. 2015;7:283ra53.
    1. Garofalo A, et al. The impact of tumor profiling approaches and genomic data strategies for cancer precision medicine. Genome Med. 2016;8:79.
    1. Schrader KA, et al. Germline Variants in Targeted Tumor Sequencing Using Matched Normal DNA. JAMA Oncol. 2016;2:104–11.
    1. Li H, Durbin R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics. 2009;25:1754–60.
    1. McKenna A, et al. The Genome Analysis Toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res. 2010;20:1297–303.
    1. Mose LE, Wilkerson MD, Hayes DN, Perou CM, Parker JS. ABRA: improved coding indel detection via assembly-based realignment. Bioinformatics. 2014;30:2813–5.
    1. Cibulskis K, et al. Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples. Nat Biotechnol. 2013;31:213–9.
    1. Ye K, Schulz MH, Long Q, Apweiler R, Ning Z. Pindel: a pattern growth approach to detect break points of large deletions and medium sized insertions from paired-end short reads. Bioinformatics. 2009;25:2865–71.
    1. Rausch T, et al. DELLY: structural variant discovery by integrated paired-end and split-read analysis. Bioinformatics. 2012;28:i333–i339.
    1. Thorvaldsdottir H, Robinson JT, Mesirov JP. Integrative Genomics Viewer (IGV): high-performance genomics data visualization and exploration. Brief Bioinform. 2013;14:178–92.
    1. Yoshihara K, et al. The landscape and therapeutic relevance of cancer-associated transcript fusions. Oncogene. 2015;34:4845–54.
    1. Manning G, Whyte DB, Martinez R, Hunter T, Sudarsanam S. The protein kinase complement of the human genome. Science. 2002;298:1912–34.
    1. Karolchik D, et al. The UCSC Table Browser data retrieval tool. Nucleic Acids Res. 2004;32:D493–6.
    1. Zheng Z, et al. Anchored multiplex PCR for targeted next-generation sequencing. Nat Med. 2014;20:1479–84.

Source: PubMed

3
Abonnieren