Network


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

Hotspot


Dive into the research topics where Zachary J. Heins is active.

Publication


Featured researches published by Zachary J. Heins.


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.


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.


JCO Precision Oncology | 2017

Prospective Genomic Profiling of Prostate Cancer Across Disease States Reveals Germline and Somatic Alterations That May Affect Clinical Decision Making

Wassim Abida; Joshua Armenia; Anuradha Gopalan; Ryan Brennan; Michael D. Walsh; David Barron; Daniel C. Danila; Dana E. Rathkopf; Michael J. Morris; Susan F. Slovin; Brigit McLaughlin; Kristen Rebecca Curtis; David M. Hyman; Jeremy C. Durack; Stephen B. Solomon; Maria E. Arcila; Ahmet Zehir; Aijazuddin Syed; Jianjiong Gao; Debyani Chakravarty; Hebert Alberto Vargas; Mark E. Robson; Joseph Vijai; Kenneth Offit; Mark T.A. Donoghue; Adam Abeshouse; Ritika Kundra; Zachary J. Heins; Alexander Penson; Christopher C. Harris

PURPOSE A long natural history and a predominant osseous pattern of metastatic spread are impediments to the adoption of precision medicine in patients with prostate cancer. To establish the feasibility of clinical genomic profiling in the disease, we performed targeted deep sequencing of tumor and normal DNA from patients with locoregional, metastatic non-castrate, and metastatic castration-resistant prostate cancer (CRPC). METHODS Patients consented to genomic analysis of their tumor and germline DNA. A hybridization capture-based clinical assay was employed to identify single nucleotide variations, small insertions and deletions, copy number alterations and structural rearrangements in over 300 cancer-related genes in tumors and matched normal blood. RESULTS We successfully sequenced 504 tumors from 451 patients with prostate cancer. Potentially actionable alterations were identified in DNA damage repair (DDR), PI3K, and MAP kinase pathways. 27% of patients harbored a germline or a somatic alteration in a DDR gene that may predict for response to PARP inhibition. Profiling of matched tumors from individual patients revealed that somatic TP53 and BRCA2 alterations arose early in tumors from patients who eventually developed metastatic disease. In contrast, comparative analysis across disease states revealed that APC alterations were enriched in metastatic tumors, while ATM alterations were specifically enriched in CRPC. CONCLUSION Through genomic profiling of prostate tumors representing the disease clinical spectrum, we identified a high frequency of potentially actionable alterations and possible drivers of disease initiation, metastasis and castration-resistance. Our findings support the routine use of tumor and germline DNA profiling for patients with advanced prostate cancer, for the purpose of guiding enrollment in targeted clinical trials and counseling families at increased risk of malignancy.


Cancer Research | 2016

Abstract 5277: The cBioPortal for cancer genomics and its application in precision oncology

Jianjiong Gao; James Lindsay; Stuart Watt; Istemi Bahceci; Pieter Lukasse; Adam Abeshouse; Hsiao-Wei Chen; Ino de Bruijn; Benjamin E. Gross; Dong Li; Ritika Kundra; Zachary J. Heins; Jorge S. Reis-Filho; Onur Sumer; Yichao Sun; Jiaojiao Wang; Qingguo Wang; Hongxin Zhang; Priti Kumari; M. Furkan Sahin; Sander de Ridder; Fedde Schaeffer; Kees van Bochove; Ugur Dogrusoz; Trevor J. Pugh; Chris Sander; Ethan Cerami; Nikolaus Schultz

The cBioPortal for Cancer Genomics provides intuitive visualization and analysis of complex cancer genomics data. The public site (http://cbioportal.org/) is accessed by more than 1,500 researchers per day, and there are now dozens of local instances of the software that host private data sets at cancer centers around the globe. We have recently released the software under an open source license, making it free to use and modify by anybody. The software and detailed documentation are available at https://github.com/cBioPortal/cbioportal. We are now establishing a multi-institutional software development network, which will coordinate and drive the future development of the software and associated data pipelines. This group will focus on four main areas: 1. New analysis and visualization features, including: a. Improved support for cross-cancer queries and cohort comparisons. b. Enhanced clinical decision support for precision oncology, including an improved patient view with knowledge base integration, patient timelines and improved tools for visualizing tumor evolution. 2. New data pipelines, including support for new genomic data types and streamlined pipelines for TCGA and the International Cancer Genome Consortium (ICGC). 3. Software architecture and performance improvements. 4. Community engagement: Documentation, user support, and training. This coordinated effort will help to further establish the cBioPortal as the software of choice in cancer genomics research, both in academia and the pharmaceutical industry. Furthermore, as the sequencing of tumor samples has entered clinical practice, we are expanding the features of the software so that it can be used for precision medicine at cancer centers. In particular, clean, web-accessible, interactive clinical reports integrating multiple sources of genome variation and clinical annotation over time has potential to improve clinical action beyond current text-based molecular reports. By making complex genomic data easily interpretable and linking it to information about drugs and clinical trials, the cBioPortal software has the potential to facilitate the use of genomic data in clinical decision making. Citation Format: Jianjiong Gao, James Lindsay, Stuart Watt, Istemi Bahceci, Pieter Lukasse, Adam Abeshouse, Hsiao-Wei Chen, Ino de Bruijn, Benjamin Gross, Dong Li, Ritika Kundra, Zachary Heins, Jorge Reis-Filho, Onur Sumer, Yichao Sun, Jiaojiao Wang, Qingguo Wang, Hongxin Zhang, Priti Kumari, M. Furkan Sahin, Sander de Ridder, Fedde Schaeffer, Kees van Bochove, Ugur Dogrusoz, Trevor Pugh, Chris Sander, Ethan Cerami, Nikolaus Schultz. The cBioPortal for cancer genomics and its application in precision oncology. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 5277.


bioRxiv | 2018

Integration and analysis of CPTAC proteomics data in the context of cancer genomics in the cBioPortal

Pamela Wu; Zachary J. Heins; James T Muller; Adam Abeshouse; Yichao Sun; Nikolaus Schultz; David Fenyö; Jianjiong Gao

The Clinical Proteomic Tumor Analysis Consortium (CPTAC) has produced extensive mass spectrometry based proteomics data for selected breast, colon and ovarian tumors from The Cancer Genome Atlas (TCGA). We have incorporated the CPTAC proteomics data into the cBioPotal to support easy exploration and integrative analysis of these proteomic datasets in the context of the clinical and genomics data from the same tumors. cBioPortal is an open source platform for exploring, visualizing, and analyzing multi-dimensional cancer genomics and clinical data. The public instance of the cBioPortal (http://cbioportal.org/) hosts more than 100 cancer genomics studies including all of the data from TCGA. Its biologist-friendly interface provides many rich analysis features, including a graphical summary of gene-level data across multiple platforms, correlation analysis between genes or other data types, survival analysis, and network visualization. Here, we present the integration of the CPTAC mass spectrometry based proteomics data into the cBioPortal, consisting of 77 breast, 95 colorectal, and 174 ovarian tumors that already have been profiled by TCGA for mutations, copy number alterations, gene expression, and DNA methylation. As a result, the CPTAC data can now be easily explored and analyzed in the cBioPortal in the context of clinical and genomics data. By integrating CPTAC data into cBioPortal, limitations of TCGA proteomics array data can be overcome while also providing a user-friendly web interface, a web API and an R client to query the mass spectrometry data together with genomic, epigenomic, and clinical data.


Cancer Research | 2017

Abstract 2607: The cBioPortal for Cancer Genomics: an open source platform for accessing and interpreting complex cancer genomics data in the era of precision medicine

Jianjiong Gao; Ersin Ciftci; Pichai Raman; Pieter Lukasse; Istemi Bahceci; Adam Abeshouse; Hsiao-Wei Chen; Ino de Bruijn; Benjamin E. Gross; Zachary J. Heins; Ritika Kundra; Aaron Lisman; Angelica Ochoa; Robert L. Sheridan; Onur Sumer; Yichao Sun; Jiaojiao Wang; Manda Wilson; Hongxin Zhang; James Xu; Andy Dufilie; Priti Kumari; James Lindsay; Anthony Cros; Karthik Kalletla; Fedde Schaeffer; Sander Tan; Sjoerd van Hagen; Jorge S. Reis-Filho; Kees van Bochove

The cBioPortal for Cancer Genomics is an open-access portal (http://cbioportal.org) that enables interactive, exploratory analysis of large-scale cancer genomics data. It integrates genomic and clinical data, and provides a suite of visualization and analysis options, including cohort and patient-level visualization, mutation visualization, survival analysis, enrichment analysis, and network analysis. The user interface is user-friendly, responsive, and makes genomic data easily accessible to translational scientists, biologists, and clinicians. The cBioPortal is a fully open source platform. All code is available on GitHub (https://github.com/cBioPortal/) under GNU Affero GPL license. The code base is maintained by multiple groups, including Memorial Sloan Kettering Cancer Center, Dana-Farber Cancer Institute, Children’s Hospital of Philadelphia, Princess Margaret Cancer Centre, and The Hyve, an open source bioinformatics company based in the Netherlands. More than 30 academic centers as well as multiple pharmaceutical and biotech companies maintain private instances of the cBioPortal. This includes the recently launched cBioPortal instance at the NCI Genomic Data Commons (https://cbioportal.gdc.nci.nih.gov/), and two large cBioPortal instances hosting genomic and clinical data at MSK and DFCI, supporting the MSK-IMPACT and DFCI Profile projects, two of the largest clinical sequencing efforts in the world. Our multi-institutional software team has accelerated the progress of evolving the core architectural technologies and developing new features to keep pace with the rapidly advancing fields of cancer genomics and precision cancer medicine. For example, we have integrated multi-platform genomics data with extensive clinical data including patient demographics, treatment history, and survival data. We have also developed a patient-centric view that visualizes both clinical and genomic data with annotation from OncoKB knowledge base. In the next few years, the development team will focus on the following areas: (1) Implementing major architectural changes to ensure future scalability and performance. (2) New features to support precision medicine, including (i) improved integration of knowledge base annotation, (ii) enhanced visualization of patient timeline, drug response, and tumor evolution, (iii) new patient similarity metrics, (iv) improved support for immunogenomics and immunotherapy, and (v) new visualization and analysis features for understanding response to therapy. (3) New analysis and target discovery features for large cohorts, including (i) supporting user-defined virtual cohort by selecting samples from multiple studies, and (ii) comparison of genomic or clinical characteristics of two or more selected cohorts. (4) Expanding community outreach, user support and training, and documentation. Citation Format: Jianjiong Gao, Ersin Ciftci, Pichai Raman, Pieter Lukasse, Istemi Bahceci, Adam Abeshouse, Hsiao-Wei Chen, Ino de Bruijn, Benjamin Gross, Zachary Heins, Ritika Kundra, Aaron Lisman, Angelica Ochoa, Robert Sheridan, Onur Sumer, Yichao Sun, Jiaojiao Wang, Manda Wilson, Hongxin Zhang, James Xu, Andy Dufilie, Priti Kumari, James Lindsay, Anthony Cros, Karthik Kalletla, Fedde Schaeffer, Sander Tan, Sjoerd van Hagen, Jorge Reis-Filho, Kees van Bochove, Ugur Dogrusoz, Trevor Pugh, Adam Resnick, Chris Sander, Ethan Cerami, Nikolaus Schultz. The cBioPortal for Cancer Genomics: an open source platform for accessing and interpreting complex cancer genomics data in the era of precision medicine [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 2607. doi:10.1158/1538-7445.AM2017-2607


Gastroenterology | 2016

Genomic Alterations Observed in Colitis-Associated Cancers Are Distinct From Those Found in Sporadic Colorectal Cancers and Vary by Type of Inflammatory Bowel Disease

Rona Yaeger; Manish A. Shah; Vincent A. Miller; Judith R. Kelsen; Kai Wang; Zachary J. Heins; Jeffrey S. Ross; Yuting He; Eric M. Sanford; Rhonda K. Yantiss; Sohail Balasubramanian; Philip J. Stephens; Nikolaus Schultz; Moshe Oren; Laura H. Tang; David P. Kelsen


Cancer Research | 2018

Abstract 3302: The molecular landscape of oncogenic signaling pathways in The Cancer Genome Atlas

Francisco Sanchez-Vega; Marco Mina; Joshua Armenia; Walid K. Chatila; Augustin Luna; Konnor La; Sofia Dimitriadoy; David Liu; Havish S. Kantheti; Zachary J. Heins; Angelica Ochoa; Benjamin E. Gross; Jianjiong Gao; Hongxin Zhang; Ritika Kundra; Cyriac Kandoth; Istemi Bahceci; Leonard Dervishi; Ugur Dogrusoz; Wanding Zhou; Hui Shen; Peter W. Laird; Alice H. Berger; Trever G. Bivona; Alexander J. Lazar; Gary D. Hammer; Thomas J. Giordano; Lawrence Kwong; Grant A. McArthur; Chenfei Huang


Journal of Clinical Oncology | 2017

Correlation of benefit from immune checkpoint inhibitors with next gen sequencing (NGS) profiles in esophagogastric cancer (EGC) patients.

Geoffrey Y. Ku; Francisco Sanchez-Vega; Walid K. Chatila; Matthew Margolis; Carly Fein; David H. Ilson; Jaclyn F. Hechtman; Yaelle Tuvy; Nancy Bouvier; Ritika Kundra; Jinru Shia; Zachary J. Heins; Sumit Middha; Ahmet Zehir; Marc Ladanyi; Michael F. Berger; Laura H. Tang; David B. Solit; Nikolaus Schultz; Yelena Y. Janjigian


Journal of Clinical Oncology | 2016

Identification of potentially actionable molecular alterations in advanced hepatocellular carcinoma (HCC).

James J. Harding; Zachary J. Heins; Louise C. Connell; Tara Soumerai; Nancy E. Kemeny; William R. Jarnagin; Eileen Mary O'Reilly; Ronald P. DeMatteo; Michael I. D'Angelica; Maeve Aine Lowery; T. Peter Kingham; Maya Gambarin; David M. Hyman; Michele Ly; Casey R. Hamilton; Michael F. Berger; David B. Solit; Leonard Saltz; Nikolaus Schultz; Ghassan K. Abou-Alfa

Collaboration


Dive into the Zachary J. Heins's collaboration.

Top Co-Authors

Avatar

Jianjiong Gao

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

Nikolaus Schultz

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Adam Abeshouse

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

Benjamin E. Gross

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

Hongxin Zhang

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

Sumit Middha

Memorial Sloan Kettering Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge