Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Walid K. Chatila is active.

Publication


Featured researches published by Walid K. Chatila.


Journal of Clinical Oncology | 2018

Molecular Determinants of Response to Anti-Programmed Cell Death (PD)-1 and Anti-Programmed Death-Ligand 1 (PD-L1) Blockade in Patients With Non-Small-Cell Lung Cancer Profiled With Targeted Next-Generation Sequencing.

Hira Rizvi; Francisco Sanchez-Vega; Konnor La; Walid K. Chatila; Philip Jonsson; Darragh Halpenny; Andrew J. Plodkowski; Niamh Long; Jennifer Sauter; Natasha Rekhtman; Travis J. Hollmann; Kurt A. Schalper; Justin F. Gainor; Ronglai Shen; Ai Ni; Kathryn Cecilia Arbour; Taha Merghoub; Jedd D. Wolchok; Alexandra Snyder; Jamie E. Chaft; Mark G. Kris; Charles M. Rudin; Nicholas D. Socci; Michael F. Berger; Barry S. Taylor; Ahmet Zehir; David B. Solit; Maria E. Arcila; Marc Ladanyi; Gregory J. Riely

Purpose Treatment of advanced non-small-cell lung cancer with immune checkpoint inhibitors (ICIs) is characterized by durable responses and improved survival in a subset of patients. Clinically available tools to optimize use of ICIs and understand the molecular determinants of response are needed. Targeted next-generation sequencing (NGS) is increasingly routine, but its role in identifying predictors of response to ICIs is not known. Methods Detailed clinical annotation and response data were collected for patients with advanced non-small-cell lung cancer treated with anti-programmed death-1 or anti-programmed death-ligand 1 [anti-programmed cell death (PD)-1] therapy and profiled by targeted NGS (MSK-IMPACT; n = 240). Efficacy was assessed by Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1, and durable clinical benefit (DCB) was defined as partial response/stable disease that lasted > 6 months. Tumor mutation burden (TMB), fraction of copy number-altered genome, and gene alterations were compared among patients with DCB and no durable benefit (NDB). Whole-exome sequencing (WES) was performed for 49 patients to compare quantification of TMB by targeted NGS versus WES. Results Estimates of TMB by targeted NGS correlated well with WES (ρ = 0.86; P < .001). TMB was greater in patients with DCB than with NDB ( P = .006). DCB was more common, and progression-free survival was longer in patients at increasing thresholds above versus below the 50th percentile of TMB (38.6% v 25.1%; P < .001; hazard ratio, 1.38; P = .024). The fraction of copy number-altered genome was highest in those with NDB. Variants in EGFR and STK11 associated with a lack of benefit. TMB and PD-L1 expression were independent variables, and a composite of TMB plus PD-L1 further enriched for benefit to ICIs. Conclusion Targeted NGS accurately estimates TMB and elevated TMB further improved likelihood of benefit to ICIs. TMB did not correlate with PD-L1 expression; both variables had similar predictive capacity. The incorporation of both TMB and PD-L1 expression into multivariable predictive models should result in greater predictive power.


Nature Genetics | 2018

The long tail of oncogenic drivers in prostate cancer.

Joshua Armenia; Stephanie A. Wankowicz; David R. Liu; Jianjiong Gao; Ritika Kundra; Ed Reznik; Walid K. Chatila; Debyani Chakravarty; G. Celine Han; Ilsa Coleman; Bruce Montgomery; Colin C. Pritchard; Colm Morrissey; Christopher E. Barbieri; Himisha Beltran; Andrea Sboner; Zafeiris Zafeiriou; Susana Miranda; Craig M. Bielski; Alexander Penson; Charlotte Tolonen; Franklin W. Huang; Dan R. Robinson; Yi Mi Wu; Robert J. Lonigro; Levi A. Garraway; Francesca Demichelis; Philip W. Kantoff; Mary-Ellen Taplin; Wassim Abida

Comprehensive genomic characterization of prostate cancer has identified recurrent alterations in genes involved in androgen signaling, DNA repair, and PI3K signaling, among others. However, larger and uniform genomic analysis may identify additional recurrently mutated genes at lower frequencies. Here we aggregate and uniformly analyze exome sequencing data from 1,013 prostate cancers. We identify and validate a new class of E26 transformation-specific (ETS)-fusion-negative tumors defined by mutations in epigenetic regulators, as well as alterations in pathways not previously implicated in prostate cancer, such as the spliceosome pathway. We find that the incidence of significantly mutated genes (SMGs) follows a long-tail distribution, with many genes mutated in less than 3% of cases. We identify a total of 97 SMGs, including 70 not previously implicated in prostate cancer, such as the ubiquitin ligase CUL3 and the transcription factor SPEN. Finally, comparing primary and metastatic prostate cancer identifies a set of genomic markers that may inform risk stratification.Meta-analysis of exome sequencing data identifies new recurrently mutated driver genes for prostate cancer. Comparison of primary and metastatic tumors further identifies genomic markers for advanced prostate cancer that may inform risk stratification.


Cancer Discovery | 2018

Genetic Predictors of Response to Systemic Therapy in Esophagogastric Cancer

Yelena Y. Janjigian; Francisco Sanchez-Vega; Philip Jonsson; Walid K. Chatila; Jaclyn F. Hechtman; Geoffrey Y. Ku; Jamie Riches; Yaelle Tuvy; Ritika Kundra; Nancy Bouvier; Efsevia Vakiani; Jianjiong Gao; Zachary J. Heins; Benjamin E. Gross; David P. Kelsen; Liying Zhang; Vivian E. Strong; Mark A. Schattner; Hans Gerdes; Daniel G. Coit; Manjit S. Bains; Zsofia K. Stadler; Valerie W. Rusch; David R. Jones; Daniela Molena; Jinru Shia; Mark E. Robson; Marinela Capanu; Sumit Middha; Ahmet Zehir

The incidence of esophagogastric cancer is rapidly rising, but only a minority of patients derive durable benefit from current therapies. Chemotherapy as well as anti-HER2 and PD-1 antibodies are standard treatments. To identify predictive biomarkers of drug sensitivity and mechanisms of resistance, we implemented prospective tumor sequencing of patients with metastatic esophagogastric cancer. There was no association between homologous recombination deficiency defects and response to platinum-based chemotherapy. Patients with microsatellite instability-high tumors were intrinsically resistant to chemotherapy but more likely to achieve durable responses to immunotherapy. The single Epstein-Barr virus-positive patient achieved a durable, complete response to immunotherapy. The level of ERBB2 amplification as determined by sequencing was predictive of trastuzumab benefit. Selection for a tumor subclone lacking ERBB2 amplification, deletion of ERBB2 exon 16, and comutations in the receptor tyrosine kinase, RAS, and PI3K pathways were associated with intrinsic and/or acquired trastuzumab resistance. Prospective genomic profiling can identify patients most likely to derive durable benefit to immunotherapy and trastuzumab and guide strategies to overcome drug resistance.Significance: Clinical application of multiplex sequencing can identify biomarkers of treatment response to contemporary systemic therapies in metastatic esophagogastric cancer. This large prospective analysis sheds light on the biological complexity and the dynamic nature of therapeutic resistance in metastatic esophagogastric cancers. Cancer Discov; 8(1); 49-58. ©2017 AACR.See related commentary by Sundar and Tan, p. 14See related article by Pectasides et al., p. 37This article is highlighted in the In This Issue feature, p. 1.


Cancer Cell | 2018

Clinical Sequencing Defines the Genomic Landscape of Metastatic Colorectal Cancer

Rona Yaeger; Walid K. Chatila; Marla Lipsyc; Jaclyn F. Hechtman; Andrea Cercek; Francisco Sanchez-Vega; Gowtham Jayakumaran; Sumit Middha; Ahmet Zehir; Mark T.A. Donoghue; Daoqi You; Agnes Viale; Nancy E. Kemeny; Neil Howard Segal; Zsofia K. Stadler; Anna M. Varghese; Ritika Kundra; Jianjiong Gao; Aijazuddin Syed; David M. Hyman; Efsevia Vakiani; Neal Rosen; Barry S. Taylor; Marc Ladanyi; Michael F. Berger; David B. Solit; Jinru Shia; Leonard Saltz; Nikolaus Schultz

Metastatic colorectal cancers (mCRCs) are clinically heterogeneous, but the genomic basis of this variability remains poorly understood. We performed prospective targeted sequencing of 1,134 CRCs. We identified splice alterations in intronic regions of APC and large in-frame deletions in CTNNB1, increasing oncogenic WNT pathway alterations to 96% of CRCs. Right-sided primary site in microsatellite stable mCRC was associated with shorter survival, older age at diagnosis, increased mutations, and enrichment of oncogenic alterations in KRAS, BRAF, PIK3CA, AKT1, RNF43, and SMAD4 compared with left-sided primaries. Left-sided tumors frequently had no identifiable genetic alteration in mitogenic signaling, but exhibited higher mitogenic ligand expression. Our results suggest different pathways to tumorigenesis in right- and left-sided microsatellite stable CRC that may underlie clinical differences.


Nature Genetics | 2018

Genome doubling shapes the evolution and prognosis of advanced cancers

Craig M. Bielski; Ahmet Zehir; Alexander Penson; Mark T.A. Donoghue; Walid K. Chatila; Joshua Armenia; Matthew T. Chang; Alison M. Schram; Philip Jonsson; Chaitanya Bandlamudi; Pedram Razavi; Gopa Iyer; Mark E. Robson; Zsofia K. Stadler; Nikolaus Schultz; José Baselga; David B. Solit; David M. Hyman; Michael F. Berger; Barry S. Taylor

Ploidy abnormalities are a hallmark of cancer, but their impact on the evolution and outcomes of cancers is unknown. Here, we identified whole-genome doubling (WGD) in the tumors of nearly 30% of 9,692 prospectively sequenced advanced cancer patients. WGD varied by tumor lineage and molecular subtype, and arose early in carcinogenesis after an antecedent transforming driver mutation. While associated with TP53 mutations, 46% of all WGD arose in TP53-wild-type tumors and in such cases was associated with an E2F-mediated G1 arrest defect, although neither aberration was obligate in WGD tumors. The variability of WGD across cancer types can be explained in part by cancer cell proliferation rates. WGD predicted for increased morbidity across cancer types, including KRAS-mutant colorectal cancers and estrogen receptor-positive breast cancers, independently of established clinical prognostic factors. We conclude that WGD is highly common in cancer and is a macro-evolutionary event associated with poor prognosis across cancer types.The authors identify whole-genome doubling (WGD) in 30% of ~10,000 sequenced tumors from patients with advanced cancer. WGD correlates with increased risk of death across cancer types.


Clinical Colorectal Cancer | 2018

FOLFCIS Treatment and Genomic Correlates of Response in Advanced Anal Squamous Cell Cancer

Sebastian Mondaca; Walid K. Chatila; David D. B. Bates; Jaclyn F. Hechtman; Andrea Cercek; Neil Howard Segal; Zsofia K. Stadler; Anna M. Varghese; Ritika Kundra; Marinela Capanu; Jinru Shia; Nikolaus Schultz; Leonard Saltz; Rona Yaeger

Micro‐Abstract In a series of 53 patients with advanced anal squamous cell cancer, we demonstrate that a modified 5‐fluorouracil and cisplatin schedule (FOLFCIS) with lower dose, more frequent administration of cisplatin is effective and well‐tolerated. This regimen should be considered a standard treatment option. Human papillomavirus‐negative anal squamous cell cancers were less sensitive to platinum‐based therapy and exhibited a distinct molecular profile. Background: Treatment of advanced anal squamous cell cancer (SCC) is usually with the combination of cisplatin and 5‐fluorouracil, which is associated with heterogeneous responses across patients and significant toxicity. We examined the safety and efficacy of a modified schedule, FOLFCIS (leucovorin, fluorouracil, and cisplatin), and performed an integrated clinical and genomic analysis of anal SCC. Patients and Methods: We reviewed all patients with advanced anal SCC receiving first‐line FOLFCIS chemotherapy – essentially a FOLFOX (leucovorin, fluorouracil, and oxaliplatin) schedule with cisplatin substituted for oxaliplatin – in our institution between 2007 and 2017, and performed deep sequencing to identify genomic markers of response and key genomic drivers. Results: Fifty‐three patients with advanced anal SCC (48 metastatic; 5 unresectable, locally advanced) received first‐line FOLFCIS during this period; all were platinum‐naive. The response rate was 48% (95% confidence interval [CI], 32.6%‐63%). With a median follow‐up of 41.6 months, progression‐free survival and overall survival were 7.1 months (95% CI, 4.4‐8.6 months) and 22.1 months (95% CI, 16.9‐28.1 months), respectively. Among all patients with advanced anal SCC that underwent sequencing during the study period, the most frequent genomic alterations consisted of chromosome 3q amplification (51%) and mutations in PIK3CA (29%) and KMT2D (22%). No genomic alteration correlated with response to platinum‐containing treatment. Although there were few cases, patients with human papillomavirus‐negative anal SCC did not appear to benefit from FOLFCIS, and all harbored distinct genomic profiles with TP53, TERT promoter, and CDKN2A mutations. Conclusions: FOLFCIS appears effective and safe as first‐line chemotherapy in patients with advanced anal SCC and represents an alternative treatment option for these patients.


Cancer Research | 2017

Abstract 4380: Integrative genomics analysis of metastatic colorectal cancer

Marla Lipsyc; Walid K. Chatila; Jaclyn F. Hechtman; Francisco Sanchez-Vega; Sumit Middha; Andrea Cercek; Zsofia K. Stadler; Ritika Kundra; Aijazuddin Syed; David M. Hyman; Ahmet Zehir; Armin Shahrokni; Anna M. Varghese; Diane Lauren Reidy; Neil Howard Segal; Efsevia Vakiani; David B. Solit; Marc Ladanyi; Michael F. Berger; Nancy E. Kemeny; Leonard Saltz; Nikolaus Schultz; Rona Yaeger

We performed an integrated clinical and bioinformatic analysis of colorectal cancers (CRCs) genotyped at our institution from 4/2014-7/2016 to comprehensively characterize genomic alterations in metastatic CRC (mCRC). We analyzed 1008 samples (474 primaries, 534 metastases) from 985 mCRC patients and 128 early stage CRCs sequenced with MSK-IMPACT, a hybridization capture next generation sequencing assay. Metastatic CRCs were divided into 3 groups by mutation burden and MSIsensor algorithm score: microsatellite stable (MSS) (n=939; 95%), microsatellite-high (MSI-H) (n=41; 4%), and ultra-mutated (n=5; 1%). Early stage CRC were enriched for MSI-H due to clinical selection and were 53% MSS, 44% MSI-H, and 4% ultra-mutated. Ultra-mutated tumors exhibited >100 mutations and harbored hotspot mutations in POLE in 8 cases and a potential novel POLE alteration in 1 case. We evaluated the frequency of oncogenic alterations in MSI-H and MSS mCRCs. The frequency of the resistance biomarkers KRAS and NRAS did not vary between MSI-H and MSS mCRCs (46% v 41%, p=0.6). Potentially actionable alterations were enriched in MSI-H tumors (78% v 33%, p Analysis of mutation frequencies in 3 MSS CRC disease states - early stage resected primary (The Cancer Genome Atlas, TCGA), primary site of mCRC, and metastatic site - found significant depletion of FBXW7 mutations in metastases. We also found significant and progressive enrichment of TP53 alterations (58% TCGA, 73% primaries of mCRC, 79% metastases) and BRAF mutations (4% TCGA, 9% primaries of mCRC, 10% metastases) in advanced disease, suggesting a role of these genes in aggressive disease. One third of the BRAF mutations in our cohorts were not V600 but known to be oncogenic. In this large dataset, we identified markers of advanced CRC and found that while MSI-H and MSS CRC have a similar frequency of resistance biomarkers, MSI-H CRC more commonly harbor actionable alterations. Citation Format: Marla Lipsyc, Walid Chatila, Jaclyn F. Hechtman, Francisco Sanchez-Vega, Sumit Middha, Andrea Cercek, Zsofia Stadler, Ritika Kundra, Aijazuddin Syed, David M. Hyman, Ahmet Zehir, Armin Shahrokni, Anna Varghese, Diane Reidy, Neil H. Segal, Efsevia Vakiani, David B. Solit, Marc Ladanyi, Michael F. Berger, Nancy Kemeny, Leonard Saltz, Nikolaus Schultz, Rona Yaeger. Integrative genomics analysis of metastatic colorectal cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 4380. doi:10.1158/1538-7445.AM2017-4380


Journal of Clinical Oncology | 2017

Molecular determinants of response and resistance to anti-PD-(L)1 blockade in patients with NSCLC profiled with targeted next-generation sequencing (NGS).

Matthew D. Hellmann; Francisco Sanchez-Vega; Konnor La; Hira Rizvi; Darragh Halpenny; Andrew J. Plodkowski; Niamh Long; Walid K. Chatila; Philip Jonsson; Kathryn Cecilia Arbour; Jamie E. Chaft; Charles M. Rudin; Mark G. Kris; Michael F. Berger; Barry S. Taylor; Ahmet Zehir; Maria E. Arcila; Marc Ladanyi; Gregory J. Riely; Nikolaus Schultz


Journal of Clinical Oncology | 2018

Tumor mutation burden (TMB) and immune-related adverse events (irAEs) compared to antibiotic (abx) use to predict for response to immune checkpoint inhibitors in esophagogastric cancer (EGC).

Megan Greally; Walid K. Chatila; Matthew Margolis; Jaclyn F. Hechtman; Yaelle Tuvy; Ritika Kundra; Marc Ladanyi; David P. Kelsen; David H. Ilson; Michael F. Berger; Laura H. Tang; David B. Solit; Nikolaus Schultz; Yelena Y. Janjigian; Geoffrey Y. Ku


Journal of Clinical Oncology | 2018

FOLFCIS regimen for treatment of cancer of the anal canal.

Sebasti´n Mondaca; Walid K. Chatila; Jaclyn F. Hechtman; David D. B. Bates; Andrea Cercek; Neil Howard Segal; Zsofia K. Stadler; Anna M. Varghese; Michael F. Berger; Marinela Capanu; Jinru Shia; Nikolaus Schultz; Leonard Saltz; Rona Yaeger

Collaboration


Dive into the Walid K. Chatila's collaboration.

Top Co-Authors

Avatar

Nikolaus Schultz

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Michael F. Berger

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Francisco Sanchez-Vega

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Jaclyn F. Hechtman

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Rona Yaeger

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

David B. Solit

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Ritika Kundra

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Zsofia K. Stadler

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Ahmet Zehir

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Jinru Shia

Memorial Sloan Kettering Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge