Network


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

Hotspot


Dive into the research topics where Matthew T. Chang is active.

Publication


Featured researches published by Matthew T. Chang.


Nature Medicine | 2017

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

Ahmet Zehir; Ryma Benayed; Ronak Shah; Aijazuddin Syed; Sumit Middha; Hyunjae R. Kim; Preethi Srinivasan; Jianjiong Gao; Debyani Chakravarty; Sean M. Devlin; Matthew D. Hellmann; David Barron; Alison M. Schram; Meera Hameed; Snjezana Dogan; Dara S. Ross; Jaclyn F. Hechtman; Deborah DeLair; Jinjuan Yao; Diana Mandelker; Donavan T. Cheng; Raghu Chandramohan; Abhinita Mohanty; Ryan Ptashkin; Gowtham Jayakumaran; Meera Prasad; Mustafa H Syed; Anoop Balakrishnan Rema; Zhen Y Liu; Khedoudja Nafa

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.


Nature Biotechnology | 2016

Identifying recurrent mutations in cancer reveals widespread lineage diversity and mutational specificity

Matthew T. Chang; Saurabh Asthana; Sizhi Paul Gao; Byron H. Lee; Jocelyn S. Chapman; Cyriac Kandoth; Jianjiong Gao; Nicholas D. Socci; David B. Solit; Adam B. Olshen; Nikolaus Schultz; Barry S. Taylor

Mutational hotspots indicate selective pressure across a population of tumor samples, but their prevalence within and across cancer types is incompletely characterized. An approach to detect significantly mutated residues, rather than methods that identify recurrently mutated genes, may uncover new biologically and therapeutically relevant driver mutations. Here, we developed a statistical algorithm to identify recurrently mutated residues in tumor samples. We applied the algorithm to 11,119 human tumors, spanning 41 cancer types, and identified 470 somatic substitution hotspots in 275 genes. We find that half of all human tumors possess one or more mutational hotspots with widespread lineage-, position- and mutant allele–specific differences, many of which are likely functional. In total, 243 hotspots were novel and appeared to affect a broad spectrum of molecular function, including hotspots at paralogous residues of Ras-related small GTPases RAC1 and RRAS2. Redefining hotspots at mutant amino acid resolution will help elucidate the allele-specific differences in their function and could have important therapeutic implications.


JCO Precision Oncology | 2017

OncoKB: A Precision Oncology Knowledge Base

Debyani Chakravarty; Jianjiong Gao; Sarah Phillips; Ritika Kundra; Hongxin Zhang; Jiaojiao Wang; Julia E. Rudolph; Rona Yaeger; Tara Soumerai; Moriah H. Nissan; Matthew T. Chang; Sarat Chandarlapaty; Tiffany A. Traina; Paul K. Paik; Alan L. Ho; Feras M. Hantash; Andrew Grupe; Shrujal S. Baxi; Margaret K. Callahan; Alexandra Snyder; Ping Chi; Daniel C. Danila; Mrinal M. Gounder; James J. Harding; Matthew D. Hellmann; Gopa Iyer; Yelena Y. Janjigian; Thomas Kaley; Douglas A. Levine; Maeve Aine Lowery

PURPOSE With prospective clinical sequencing of tumors emerging as a mainstay in cancer care, there is an urgent need for a clinical support tool that distills the clinical implications associated with specific mutation events into a standardized and easily interpretable format. To this end, we developed OncoKB, an expert-guided precision oncology knowledge base. METHODS OncoKB annotates the biological and oncogenic effect and the prognostic and predictive significance of somatic molecular alterations. Potential treatment implications are stratified by the level of evidence that a specific molecular alteration is predictive of drug response based on US Food and Drug Administration (FDA) labeling, National Comprehensive Cancer Network (NCCN) guidelines, disease-focused expert group recommendations and the scientific literature. RESULTS To date, over 3000 unique mutations, fusions, and copy number alterations in 418 cancer-associated genes have been annotated. To test the utility of OncoKB, we annotated all genomic events in 5983 primary tumor samples in 19 cancer types. Forty-one percent of samples harbored at least one potentially actionable alteration, of which 7.5% were predictive of clinical benefit from a standard treatment. OncoKB annotations are available through a public web resource (http://oncokb.org/) and are also incorporated into the cBioPortal for Cancer Genomics to facilitate the interpretation of genomic alterations by physicians and researchers. CONCLUSION OncoKB, a comprehensive and curated precision oncology knowledge base, offers oncologists detailed, evidence-based information about individual somatic mutations and structural alterations present in patient tumors with the goal of supporting optimal treatment decisions.


Nature Genetics | 2016

Recurrent activating mutations of G-protein-coupled receptor CYSLTR2 in uveal melanoma

Amanda R. Moore; Emilie Ceraudo; Jessica Sher; Youxin Guan; Alexander N. Shoushtari; Matthew T. Chang; Jenny Zhang; Edward Walczak; Manija A. Kazmi; Barry S. Taylor; Thomas Huber; Ping Chi; Thomas P. Sakmar; Yu Chen

Uveal melanomas are molecularly distinct from cutaneous melanomas and lack mutations in BRAF, NRAS, KIT, and NF1. Instead, they are characterized by activating mutations in GNAQ and GNA11, two highly homologous α subunits of Gαq/11 heterotrimeric G proteins, and in PLCB4 (phospholipase C β4), the downstream effector of Gαq signaling. We analyzed genomics data from 136 uveal melanoma samples and found a recurrent mutation in CYSLTR2 (cysteinyl leukotriene receptor 2) encoding a p.Leu129Gln substitution in 4 of 9 samples that lacked mutations in GNAQ, GNA11, and PLCB4 but in 0 of 127 samples that harbored mutations in these genes. The Leu129Gln CysLT2R mutant protein constitutively activates endogenous Gαq and is unresponsive to stimulation by leukotriene. Expression of Leu129Gln CysLT2R in melanocytes enforces expression of a melanocyte-lineage signature, drives phorbol ester–independent growth in vitro, and promotes tumorigenesis in vivo. Our findings implicate CYSLTR2 as a uveal melanoma oncogene and highlight the critical role of Gαq signaling in uveal melanoma pathogenesis.


Cancer Discovery | 2017

Prospective Comprehensive Molecular Characterization of Lung Adenocarcinomas for Efficient Patient Matching to Approved and Emerging Therapies

Emmet Jordan; Hyunjae R. Kim; Maria E. Arcila; David Barron; Debyani Chakravarty; Jianjiong Gao; Matthew T. Chang; Andy Ni; Ritika Kundra; Philip Jonsson; Gowtham Jayakumaran; Sizhi Paul Gao; Hannah Johnsen; Aphrothiti J. Hanrahan; Ahmet Zehir; Natasha Rekhtman; Michelle S. Ginsberg; Bob T. Li; Helena A. Yu; Paul K. Paik; Alexander Drilon; Matthew D. Hellmann; Dalicia Reales; Ryma Benayed; Valerie W. Rusch; Mark G. Kris; Jamie E. Chaft; José Baselga; Barry S. Taylor; Nikolaus Schultz

Tumor genetic testing is standard of care for patients with advanced lung adenocarcinoma, but the fraction of patients who derive clinical benefit remains undefined. Here, we report the experience of 860 patients with metastatic lung adenocarcinoma analyzed prospectively for mutations in >300 cancer-associated genes. Potentially actionable genetic events were stratified into one of four levels based upon published clinical or laboratory evidence that the mutation in question confers increased sensitivity to standard or investigational therapies. Overall, 37.1% (319/860) of patients received a matched therapy guided by their tumor molecular profile. Excluding alterations associated with standard-of-care therapy, 14.4% (69/478) received matched therapy, with a clinical benefit of 52%. Use of matched therapy was strongly influenced by the level of preexistent clinical evidence that the mutation identified predicts for drug response. Analysis of genes mutated significantly more often in tumors without known actionable mutations nominated STK11 and KEAP1 as possible targetable mitogenic drivers.Significance: An increasing number of therapies that target molecular alterations required for tumor maintenance and progression have demonstrated clinical activity in patients with lung adenocarcinoma. The data reported here suggest that broader, early testing for molecular alterations that have not yet been recognized as standard-of-care predictive biomarkers of drug response could accelerate the development of targeted agents for rare mutational events and could result in improved clinical outcomes. Cancer Discov; 7(6); 596-609. ©2017 AACR.See related commentary by Liu et al., p. 555This article is highlighted in the In This Issue feature, p. 539.


Nature | 2017

Tumours with class 3 BRAF mutants are sensitive to the inhibition of activated RAS

Zhan Yao; Rona Yaeger; Vanessa Rodrik-Outmezguine; Anthony Tao; Neilawattie M. Torres; Matthew T. Chang; Matthias Drosten; Huiyong Zhao; Fabiola Cecchi; Todd Hembrough; Judith Michels; H. Baumert; Linde A. Miles; Naomi M. Campbell; Elisa de Stanchina; David B. Solit; Mariano Barbacid; Barry S. Taylor; Neal Rosen

Approximately 200 BRAF mutant alleles have been identified in human tumours. Activating BRAF mutants cause feedback inhibition of GTP-bound RAS, are RAS-independent and signal either as active monomers (class 1) or constitutively active dimers (class 2). Here we characterize a third class of BRAF mutants—those that have impaired kinase activity or are kinase-dead. These mutants are sensitive to ERK-mediated feedback and their activation of signalling is RAS-dependent. The mutants bind more tightly than wild-type BRAF to RAS–GTP, and their binding to and activation of wild-type CRAF is enhanced, leading to increased ERK signalling. The model suggests that dysregulation of signalling by these mutants in tumours requires coexistent mechanisms for maintaining RAS activation despite ERK-dependent feedback. Consistent with this hypothesis, melanomas with these class 3 BRAF mutations also harbour RAS mutations or NF1 deletions. By contrast, in lung and colorectal cancers with class 3 BRAF mutants, RAS is typically activated by receptor tyrosine kinase signalling. These tumours are sensitive to the inhibition of RAS activation by inhibitors of receptor tyrosine kinases. We have thus defined three distinct functional classes of BRAF mutants in human tumours. The mutants activate ERK signalling by different mechanisms that dictate their sensitivity to therapeutic inhibitors of the pathway.


Journal of Clinical Oncology | 2017

AKT Inhibition in Solid Tumors With AKT1 Mutations

David M. Hyman; Lillian Mary Smyth; Mark T.A. Donoghue; Shannon N. Westin; Philippe L. Bedard; Emma Dean; H Bando; Anthony B. El-Khoueiry; J. A Perez-Fidalgo; A Mita; Jan H. M. Schellens; Matthew T. Chang; J. B Reichel; Nancy Bouvier; S. D Selcuklu; Tara Soumerai; J Torrisi; Joseph P. Erinjeri; Helen Ambrose; J. C Barrett; B Dougherty; Andrew Foxley; Justin Lindemann; R McEwen; Martin Pass; Gaia Schiavon; Michael F. Berger; Sarat Chandarlapaty; David B. Solit; Udai Banerji

Purpose AKT1 E17K mutations are oncogenic and occur in many cancers at a low prevalence. We performed a multihistology basket study of AZD5363, an ATP-competitive pan-AKT kinase inhibitor, to determine the preliminary activity of AKT inhibition in AKT-mutant cancers. Patients and Methods Fifty-eight patients with advanced solid tumors were treated. The primary end point was safety; secondary end points were progression-free survival (PFS) and response according to Response Evaluation Criteria in Solid Tumors (RECIST). Tumor biopsies and plasma cell-free DNA (cfDNA) were collected in the majority of patients to identify predictive biomarkers of response. Results In patients with AKT1 E17K-mutant tumors (n = 52) and a median of five lines of prior therapy, the median PFS was 5.5 months (95% CI, 2.9 to 6.9 months), 6.6 months (95% CI, 1.5 to 8.3 months), and 4.2 months (95% CI, 2.1 to 12.8 months) in patients with estrogen receptor-positive breast, gynecologic, and other solid tumors, respectively. In an exploratory biomarker analysis, imbalance of the AKT1 E17K-mutant allele, most frequently caused by copy-neutral loss-of-heterozygosity targeting the wild-type allele, was associated with longer PFS (hazard ratio [HR], 0.41; P = .04), as was the presence of coincident PI3K pathway hotspot mutations (HR, 0.21; P = .045). Persistent declines in AKT1 E17K in cfDNA were associated with improved PFS (HR, 0.18; P = .004) and response ( P = .025). Responses were not restricted to patients with detectable AKT1 E17K in pretreatment cfDNA. The most common grade ≥ 3 adverse events were hyperglycemia (24%), diarrhea (17%), and rash (15.5%). Conclusion This study provides the first clinical data that AKT1 E17K is a therapeutic target in human cancer. The genomic context of the AKT1 E17K mutation further conditioned response to AZD5363.


Genome Medicine | 2017

3D clusters of somatic mutations in cancer reveal numerous rare mutations as functional targets

Jianjiong Gao; Matthew T. Chang; Hannah Johnsen; Sizhi Paul Gao; Brooke E. Sylvester; Selcuk Onur Sumer; Hongxin Zhang; David B. Solit; Barry S. Taylor; Nikolaus Schultz; Chris Sander

Many mutations in cancer are of unknown functional significance. Standard methods use statistically significant recurrence of mutations in tumor samples as an indicator of functional impact. We extend such analyses into the long tail of rare mutations by considering recurrence of mutations in clusters of spatially close residues in protein structures. Analyzing 10,000 tumor exomes, we identify more than 3000 rarely mutated residues in proteins as potentially functional and experimentally validate several in RAC1 and MAP2K1. These potential driver mutations (web resources: 3dhotspots.org and cBioPortal.org) can extend the scope of genomically informed clinical trials and of personalized choice of therapy.


Nature | 2017

USP7 small-molecule inhibitors interfere with ubiquitin binding

Lorna Kategaya; Paola Di Lello; Lionel Rouge; Richard Pastor; Kevin R. Clark; Jason Drummond; Tracy Kleinheinz; Eva Lin; John-Paul Upton; Sumit Prakash; Johanna Heideker; Mark L. McCleland; Maria Stella Ritorto; Dario R. Alessi; Matthias Trost; Travis W. Bainbridge; Michael C. M. Kwok; Taylur P. Ma; Zachary Stiffler; Bradley Brasher; Yinyan Tang; Priyadarshini Jaishankar; Brian R. Hearn; Adam R. Renslo; Michelle R. Arkin; Fred E. Cohen; Kebing Yu; Frank Peale; Florian Gnad; Matthew T. Chang

The ubiquitin system regulates essential cellular processes in eukaryotes. Ubiquitin is ligated to substrate proteins as monomers or chains and the topology of ubiquitin modifications regulates substrate interactions with specific proteins. Thus ubiquitination directs a variety of substrate fates including proteasomal degradation. Deubiquitinase enzymes cleave ubiquitin from substrates and are implicated in disease; for example, ubiquitin-specific protease-7 (USP7) regulates stability of the p53 tumour suppressor and other proteins critical for tumour cell survival. However, developing selective deubiquitinase inhibitors has been challenging and no co-crystal structures have been solved with small-molecule inhibitors. Here, using nuclear magnetic resonance-based screening and structure-based design, we describe the development of selective USP7 inhibitors GNE-6640 and GNE-6776. These compounds induce tumour cell death and enhance cytotoxicity with chemotherapeutic agents and targeted compounds, including PIM kinase inhibitors. Structural studies reveal that GNE-6640 and GNE-6776 non-covalently target USP7 12 Å distant from the catalytic cysteine. The compounds attenuate ubiquitin binding and thus inhibit USP7 deubiquitinase activity. GNE-6640 and GNE-6776 interact with acidic residues that mediate hydrogen-bond interactions with the ubiquitin Lys48 side chain, suggesting that USP7 preferentially interacts with and cleaves ubiquitin moieties that have free Lys48 side chains. We investigated this idea by engineering di-ubiquitin chains containing differential proximal and distal isotopic labels and measuring USP7 binding by nuclear magnetic resonance. This preferential binding protracted the depolymerization kinetics of Lys48-linked ubiquitin chains relative to Lys63-linked chains. In summary, engineering compounds that inhibit USP7 activity by attenuating ubiquitin binding suggests opportunities for developing other deubiquitinase inhibitors and may be a strategy more broadly applicable to inhibiting proteins that require ubiquitin binding for full functional activity.


Nature Reviews Clinical Oncology | 2017

Fusions in solid tumours: diagnostic strategies, targeted therapy, and acquired resistance

Alison M. Schram; Matthew T. Chang; Philip Jonsson; Alexander Drilon

Structural gene rearrangements resulting in gene fusions are frequent events in solid tumours. The identification of certain activating fusions can aid in the diagnosis and effective treatment of patients with tumours harbouring these alterations. Advances in the techniques used to identify fusions have enabled physicians to detect these alterations in the clinic. Targeted therapies directed at constitutively activated oncogenic tyrosine kinases have proven remarkably effective against cancers with fusions involving ALK, ROS1, or PDGFB, and the efficacy of this approach continues to be explored in malignancies with RET, NTRK1/2/3, FGFR1/2/3, and BRAF/CRAF fusions. Nevertheless, prolonged treatment with such tyrosine-kinase inhibitors (TKIs) leads to the development of acquired resistance to therapy. This resistance can be mediated by mutations that alter drug binding, or by the activation of bypass pathways. Second-generation and third-generation TKIs have been developed to overcome resistance, and have variable levels of activity against tumours harbouring individual mutations that confer resistance to first-generation TKIs. The rational sequential administration of different inhibitors is emerging as a new treatment paradigm for patients with tumours that retain continued dependency on the downstream kinase of interest.

Collaboration


Dive into the Matthew T. Chang's collaboration.

Top Co-Authors

Avatar

Barry S. Taylor

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

Nikolaus Schultz

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Jianjiong Gao

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
Top Co-Authors

Avatar

Michael F. Berger

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Cyriac Kandoth

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Nicholas D. Socci

Memorial Sloan Kettering Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge