Network


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

Hotspot


Dive into the research topics where Jianjiong Gao is active.

Publication


Featured researches published by Jianjiong Gao.


Cancer Discovery | 2012

The cBio Cancer Genomics Portal: An Open Platform for Exploring Multidimensional Cancer Genomics Data

Ethan Cerami; Jianjiong Gao; Ugur Dogrusoz; Benjamin E. Gross; Selcuk Onur Sumer; Bülent Arman Aksoy; Anders Jacobsen; Caitlin J. Byrne; Michael L. Heuer; Erik G. Larsson; Yevgeniy Antipin; Boris Reva; Arthur P. Goldberg; Chris Sander; Nikolaus Schultz

The cBio Cancer Genomics Portal (http://cbioportal.org) is an open-access resource for interactive exploration of multidimensional cancer genomics data sets, currently providing access to data from more than 5,000 tumor samples from 20 cancer studies. The cBio Cancer Genomics Portal significantly lowers the barriers between complex genomic data and cancer researchers who want rapid, intuitive, and high-quality access to molecular profiles and clinical attributes from large-scale cancer genomics projects and empowers researchers to translate these rich data sets into biologic insights and clinical applications.


Cell | 2015

Integrative clinical genomics of advanced prostate cancer

Dan R. Robinson; Eliezer M. Van Allen; Yi Mi Wu; Nikolaus Schultz; Robert J. Lonigro; Juan Miguel Mosquera; Bruce Montgomery; Mary-Ellen Taplin; Colin C. Pritchard; Gerhardt Attard; Himisha Beltran; Wassim Abida; Robert K. Bradley; Jake Vinson; Xuhong Cao; Pankaj Vats; Lakshmi P. Kunju; Maha Hussain; Felix Y. Feng; Scott A. Tomlins; Kathleen A. Cooney; David C. Smith; Christine Brennan; Javed Siddiqui; Rohit Mehra; Yu Chen; Dana E. Rathkopf; Michael J. Morris; Stephen B. Solomon; Jeremy C. Durack

Toward development of a precision medicine framework for metastatic, castration-resistant prostate cancer (mCRPC), we established a multi-institutional clinical sequencing infrastructure to conduct prospective whole-exome and transcriptome sequencing of bone or soft tissue tumor biopsies from a cohort of 150 mCRPC affected individuals. Aberrations of AR, ETS genes, TP53, and PTEN were frequent (40%-60% of cases), with TP53 and AR alterations enriched in mCRPC compared to primary prostate cancer. We identified new genomic alterations in PIK3CA/B, R-spondin, BRAF/RAF1, APC, β-catenin, and ZBTB16/PLZF. Moreover, aberrations of BRCA2, BRCA1, and ATM were observed at substantially higher frequencies (19.3% overall) compared to those in primary prostate cancers. 89% of affected individuals harbored a clinically actionable aberration, including 62.7% with aberrations in AR, 65% in other cancer-related genes, and 8% with actionable pathogenic germline alterations. This cohort study provides clinically actionable information that could impact treatment decisions for these affected individuals.


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.


Nature Genetics | 2014

Recurrent SMARCA4 mutations in small cell carcinoma of the ovary

Petar Jelinic; J.J. Mueller; Narciso Olvera; Fanny Dao; Sasinya N. Scott; Ronak Shah; Jianjiong Gao; Nikolaus Schultz; Mithat Gonen; Robert A. Soslow; Michael F. Berger; Douglas A. Levine

Small cell carcinoma of the ovary, hypercalcemic type (SCCOHT) is a rare, highly aggressive form of ovarian cancer primarily diagnosed in young women. We identified inactivating biallelic SMARCA4 mutations in 100% of the 12 SCCOHT tumors examined. Protein studies confirmed loss of SMARCA4 expression, suggesting a key role for the SWI/SNF chromatin-remodeling complex in SCCOHT.


Plant Physiology | 2012

Phosphoproteomic Analysis of Seed Maturation in Arabidopsis, Rapeseed, and Soybean

Louis Meyer; Jianjiong Gao; Dong Xu; Jay J. Thelen

To characterize protein phosphorylation in developing seed, a large-scale, mass spectrometry-based phosphoproteomic study was performed on whole seeds at five sequential stages of development in soybean (Glycine max), rapeseed (Brassica napus), and Arabidopsis (Arabidopsis thaliana). Phosphopeptides were enriched from 0.5 mg of total peptides using a combined strategy of immobilized metal affinity and metal oxide affinity chromatography. Enriched phosphopeptides were analyzed by Orbitrap tandem mass spectrometry and mass spectra mined against cognate genome or cDNA databases in both forward and randomized orientations, the latter to calculate false discovery rate. We identified a total of 2,001 phosphopeptides containing 1,026 unambiguous phosphorylation sites from 956 proteins, with an average false discovery rate of 0.78% for the entire study. The entire data set was uploaded into the Plant Protein Phosphorylation Database (www.p3db.org), including all meta-data and annotated spectra. The Plant Protein Phosphorylation Database is a portal for all plant phosphorylation data and allows for homology-based querying of experimentally determined phosphosites. Comparisons with other large-scale phosphoproteomic studies determined that 652 of the phosphoproteins are novel to this study. The unique proteins fall into several Gene Ontology categories, some of which are overrepresented in our study as well as other large-scale phosphoproteomic studies, including metabolic process and RNA binding; other categories are only overrepresented in our study, like embryonic development. This investigation shows the importance of analyzing multiple plants and plant organs to comprehensively map the complete plant phosphoproteome.


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 | 2016

Deletions linked to TP53 loss drive cancer through p53-independent mechanisms

Yu Liu; Chong Chen; Zhengmin Xu; Claudio Scuoppo; Cory D. Rillahan; Jianjiong Gao; Barbara Spitzer; Benedikt Bosbach; Edward R. Kastenhuber; Timour Baslan; Sarah Ackermann; Lihua Cheng; Qingguo Wang; Ting Niu; Nikolaus Schultz; Ross L. Levine; Alea A. Mills; Scott W. Lowe

Mutations disabling the TP53 tumour suppressor gene represent the most frequent events in human cancer and typically occur through a two-hit mechanism involving a missense mutation in one allele and a ‘loss of heterozygosity’ deletion encompassing the other. While TP53 missense mutations can also contribute gain-of-function activities that impact tumour progression, it remains unclear whether the deletion event, which frequently includes many genes, impacts tumorigenesis beyond TP53 loss alone. Here we show that somatic heterozygous deletion of mouse chromosome 11B3, a 4-megabase region syntenic to human 17p13.1, produces a greater effect on lymphoma and leukaemia development than Trp53 deletion. Mechanistically, the effect of 11B3 loss on tumorigenesis involves co-deleted genes such as Eif5a and Alox15b (also known as Alox8), the suppression of which cooperates with Trp53 loss to produce more aggressive disease. Our results imply that the selective advantage produced by human chromosome 17p deletion reflects the combined impact of TP53 loss and the reduced dosage of linked tumour suppressor genes.


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.


Cell | 2018

Comprehensive Characterization of Cancer Driver Genes and Mutations

Matthew Bailey; Collin Tokheim; Eduard Porta-Pardo; Sohini Sengupta; Denis Bertrand; Amila Weerasinghe; Antonio Colaprico; Michael C. Wendl; Jaegil Kim; Brendan Reardon; Patrick Kwok Shing Ng; Kang Jin Jeong; Song Cao; Zixing Wang; Jianjiong Gao; Qingsong Gao; Fang Wang; Eric Minwei Liu; Loris Mularoni; Carlota Rubio-Perez; Niranjan Nagarajan; Isidro Cortes-Ciriano; Daniel Cui Zhou; Wen-Wei Liang; Julian Hess; Venkata Yellapantula; David Tamborero; Abel Gonzalez-Perez; Chayaporn Suphavilai; Jia Yu Ko

Identifying molecular cancer drivers is critical for precision oncology. Multiple advanced algorithms to identify drivers now exist, but systematic attempts to combine and optimize them on large datasets are few. We report a PanCancer and PanSoftware analysis spanning 9,423 tumor exomes (comprising all 33 of The Cancer Genome Atlas projects) and using 26 computational tools to catalog driver genes and mutations. We identify 299 driver genes with implications regarding their anatomical sites and cancer/cell types. Sequence- and structure-based analyses identified >3,400 putative missense driver mutations supported by multiple lines of evidence. Experimental validation confirmed 60%-85% of predicted mutations as likely drivers. We found that >300 MSI tumors are associated with high PD-1/PD-L1, and 57% of tumors analyzed harbor putative clinically actionable events. Our study represents the most comprehensive discovery of cancer genes and mutations to date and will serve as a blueprint for future biological and clinical endeavors.

Collaboration


Dive into the Jianjiong Gao's collaboration.

Top Co-Authors

Avatar

Nikolaus Schultz

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

Debyani Chakravarty

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Barry S. Taylor

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

David B. Solit

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

Michael F. Berger

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Matthew T. Chang

Memorial Sloan Kettering Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge