Network


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

Hotspot


Dive into the research topics where John Paul Shen is active.

Publication


Featured researches published by John Paul Shen.


Nature Methods | 2013

Network-based stratification of tumor mutations

Matan Hofree; John Paul Shen; Hannah Carter; Andrew M. Gross; Trey Ideker

Many forms of cancer have multiple subtypes with different causes and clinical outcomes. Somatic tumor genome sequences provide a rich new source of data for uncovering these subtypes but have proven difficult to compare, as two tumors rarely share the same mutations. Here we introduce network-based stratification (NBS), a method to integrate somatic tumor genomes with gene networks. This approach allows for stratification of cancer into informative subtypes by clustering together patients with mutations in similar network regions. We demonstrate NBS in ovarian, uterine and lung cancer cohorts from The Cancer Genome Atlas. For each tissue, NBS identifies subtypes that are predictive of clinical outcomes such as patient survival, response to therapy or tumor histology. We identify network regions characteristic of each subtype and show how mutation-derived subtypes can be used to train an mRNA expression signature, which provides similar information in the absence of DNA sequence.


Nature Genetics | 2014

Multi-tiered genomic analysis of head and neck cancer ties TP53 mutation to 3p loss

Andrew M. Gross; Ryan K. Orosco; John Paul Shen; Ann Marie Egloff; Hannah Carter; Matan Hofree; Michel Choueiri; Charles S. Coffey; Scott M. Lippman; D. N. Hayes; Ezra E.W. Cohen; Grandis; Quyen T. Nguyen; Trey Ideker

Head and neck squamous cell carcinoma (HNSCC) is characterized by aggressive behavior with a propensity for metastasis and recurrence. Here we report a comprehensive analysis of the molecular and clinical features of HNSCC that govern patient survival. We find that TP53 mutation is frequently accompanied by loss of chromosome 3p and that the combination of these events is associated with a surprising decrease in survival time (1.9 years versus >5 years for TP53 mutation alone). The TP53-3p interaction is specific to chromosome 3p and validates in HNSCC and pan-cancer cohorts. In human papillomavirus (HPV)-positive tumors, in which HPV inactivates TP53, 3p deletion is also common and is associated with poor outcomes. The TP53-3p event is modified by mir-548k expression, which decreases survival further, and is mutually exclusive with mutations affecting RAS signaling. Together, the identified markers underscore the molecular heterogeneity of HNSCC and enable a new multi-tiered classification of this disease.


Nature Methods | 2017

Combinatorial CRISPR–Cas9 screens for de novo mapping of genetic interactions

John Paul Shen; Dongxin Zhao; Roman Sasik; Jens Luebeck; Amanda Birmingham; Ana Bojorquez-Gomez; Katherine Licon; Kristin Klepper; Daniel Pekin; Alex N. Beckett; Kyle Salinas Sanchez; Alex Thomas; Chih-Chung Kuo; Dan Du; Assen Roguev; Nathan E. Lewis; Aaron N. Chang; Jason F. Kreisberg; Nevan J. Krogan; Lei S. Qi; Trey Ideker; Prashant Mali

We developed a systematic approach to map human genetic networks by combinatorial CRISPR–Cas9 perturbations coupled to robust analysis of growth kinetics. We targeted all pairs of 73 cancer genes with dual guide RNAs in three cell lines, comprising 141,912 tests of interaction. Numerous therapeutically relevant interactions were identified, and these patterns replicated with combinatorial drugs at 75% precision. From these results, we anticipate that cellular context will be critical to synthetic-lethal therapies.


Molecular Cell | 2016

A Network of Conserved Synthetic Lethal Interactions for Exploration of Precision Cancer Therapy

Rohith Srivas; John Paul Shen; Chih Cheng Yang; Su Ming Sun; Jianfeng Li; Andrew M. Gross; James Jensen; Katherine Licon; Ana Bojorquez-Gomez; Kristin Klepper; Justin K. Huang; Daniel Pekin; Jia L. Xu; Huwate Yeerna; Vignesh Sivaganesh; Leonie Kollenstart; Haico van Attikum; Pedro Aza-Blanc; Robert W. Sobol; Trey Ideker

An emerging therapeutic strategy for cancer is to induce selective lethality in a tumor by exploiting interactions between its driving mutations and specific drug targets. Here we use a multi-species approach to develop a resource of synthetic lethal interactions relevant to cancer therapy. First, we screen in yeast ∼169,000 potential interactions among orthologs of human tumor suppressor genes (TSG) and genes encoding drug targets across multiple genotoxic environments. Guided by the strongest signal, we evaluate thousands of TSG-drug combinations in HeLa cells, resulting in networks of conserved synthetic lethal interactions. Analysis of these networks reveals that interaction stability across environments and shared gene function increase the likelihood of observing an interaction in human cancer cells. Using these rules, we prioritize ∼10(5) human TSG-drug combinations for future follow-up. We validate interactions based on cell and/or patient survival, including topoisomerases with RAD17 and checkpoint kinases with BLM.


Nature Methods | 2017

Genetic interaction mapping in mammalian cells using CRISPR interference

Dan Du; Assen Roguev; David E. Gordon; Meng Chen; Si‐Han Chen; Michael Shales; John Paul Shen; Trey Ideker; Prashant Mali; Lei S. Qi; Nevan J. Krogan

We describe a combinatorial CRISPR interference (CRISPRi) screening platform for mapping genetic interactions in mammalian cells. We targeted 107 chromatin-regulation factors in human cells with pools of either single or double single guide RNAs (sgRNAs) to downregulate individual genes or gene pairs, respectively. Relative enrichment analysis of individual sgRNAs or sgRNA pairs allowed for quantitative characterization of genetic interactions, and comparison with protein–protein-interaction data revealed a functional map of chromatin regulation.


Haematologica | 2015

Oral administration of a bone morphogenetic protein type I receptor inhibitor prevents the development of anemia of inflammation.

Claire Mayeur; Starsha A. Kolodziej; Amy Wang; Xin Xu; Arthur Lee; Paul B. Yu; John Paul Shen; Kenneth D. Bloch; Donald B. Bloch

Anemia of inflammation (AI), the second most frequent form of anemia, complicates inflammatory and chronic diseases.[1][1] Although typically mild to moderate, AI is associated with increased morbidity and mortality.[1][1] One of the mechanisms contributing to the development of AI is a persistent


PLOS ONE | 2015

ERCC1 and TS Expression as Prognostic and Predictive Biomarkers in Metastatic Colon Cancer.

Michel Choueiri; John Paul Shen; Andrew M. Gross; Justin K. Huang; Trey Ideker; Paul T. Fanta

In patients with metastatic colon cancer, response to first line chemotherapy is a strong predictor of overall survival (OS). Currently, oncologists lack diagnostic tests to determine which chemotherapy regimen offers the greatest chance for response in an individual patient. Here we present the results of gene expression analysis for two genes, ERCC1 and TS, measured with the commercially available ResponseDX: Colon assay (Response Genetics, Los Angeles, CA) in 41 patients with de novo metastatic colon cancer diagnosed between July 2008 and August 2013 at the University of California, San Diego. In addition ERCC1 and TS expression levels as determined by RNAseq and survival data for patients in TCGA were downloaded from the TCGA data portal. We found that patients with low expression of ERCC1 (n = 33) had significantly longer median OS (36.0 vs. 10.1 mo, HR 0.29, 95% CI .095 to .84, log-rank p = 9.0x10-6) and median time to treatment to failure (TTF) following first line chemotherapy (14.1 vs. 2.4 mo, HR 0.17, 95% CI 0.048 to 0.58, log-rank p = 5.3x10-4) relative to those with high expression (n = 4). After accounting for the covariates age, sex, tumor grade and ECOG performance status in a Cox proportional hazard model the association of low ERCC1 with longer OS (HR 0.18, 95% CI 0.14 to 0.26, p = 0.0448) and TTF (HR 0.16, 95% CI 0.14 to 0.21, p = 0.0053) remained significant. Patients with low TS expression (n = 29) had significantly longer median OS (36.0 vs. 14.8 mo, HR 0.25, 95% CI 0.074 to 0.82, log-rank p = 0.022) relative to those with high expression (n = 12). The combined low expression of ERCC1/TS was predictive of response in patients treated with FOLFOX (40% vs. 91%, RR 2.3, Fisher’s exact test p = 0.03, n = 27), but not with FOLFIRI (71% vs. 71%, RR 1.0, Fisher’s exact test p = 1, n = 14). Overall, these findings suggest that measurement of ERCC1 and TS expression has potential clinical utility in managing patients with metastatic colorectal cancer.


Oncotarget | 2015

Chemogenetic profiling identifies RAD17 as synthetically lethal with checkpoint kinase inhibition

John Paul Shen; Rohith Srivas; Andrew M. Gross; Jianfeng Li; Eric J. Jaehnig; Su Ming Sun; Ana Bojorquez-Gomez; Katherine Licon; Vignesh Sivaganesh; Jia L. Xu; Kristin Klepper; Huwate Yeerna; Daniel Pekin; Chu Ping Qiu; Haico van Attikum; Robert W. Sobol; Trey Ideker

Chemical inhibitors of the checkpoint kinases have shown promise in the treatment of cancer, yet their clinical utility may be limited by a lack of molecular biomarkers to identify specific patients most likely to respond to therapy. To this end, we screened 112 known tumor suppressor genes for synthetic lethal interactions with inhibitors of the CHEK1 and CHEK2 checkpoint kinases. We identified eight interactions, including the Replication Factor C (RFC)-related protein RAD17. Clonogenic assays in RAD17 knockdown cell lines identified a substantial shift in sensitivity to checkpoint kinase inhibition (3.5-fold) as compared to RAD17 wild-type. Additional evidence for this interaction was found in a large-scale functional shRNA screen of over 100 genotyped cancer cell lines, in which CHEK1/2 mutant cell lines were unexpectedly sensitive to RAD17 knockdown. This interaction was widely conserved, as we found that RAD17 interacts strongly with checkpoint kinases in the budding yeast Saccharomyces cerevisiae. In the setting of RAD17 knockdown, CHEK1/2 inhibition was found to be synergistic with inhibition of WEE1, another pharmacologically relevant checkpoint kinase. Accumulation of the DNA damage marker γH2AX following chemical inhibition or transient knockdown of CHEK1, CHEK2 or WEE1 was magnified by knockdown of RAD17. Taken together, our data suggest that CHEK1 or WEE1 inhibitors are likely to have greater clinical efficacy in tumors with RAD17 loss-of-function.


Nature Genetics | 2018

A global transcriptional network connecting noncoding mutations to changes in tumor gene expression

Wei Zhang; Ana Bojorquez-Gomez; Daniel Ortiz Velez; Guorong Xu; Kyle Salinas Sanchez; John Paul Shen; Kevin N. H. Chen; Katherine Licon; Collin Melton; Katrina M. Olson; Michael Ku Yu; Justin K. Huang; Hannah Carter; Emma K. Farley; Michael Snyder; Stephanie I. Fraley; Jason F. Kreisberg; Trey Ideker

Although cancer genomes are replete with noncoding mutations, the effects of these mutations remain poorly characterized. Here we perform an integrative analysis of 930 tumor whole genomes and matched transcriptomes, identifying a network of 193 noncoding loci in which mutations disrupt target gene expression. These ‘somatic eQTLs’ (expression quantitative trait loci) are frequently mutated in specific cancer tissues, and the majority can be validated in an independent cohort of 3,382 tumors. Among these, we find that the effects of noncoding mutations on DAAM1, MTG2 and HYI transcription are recapitulated in multiple cancer cell lines and that increasing DAAM1 expression leads to invasive cell migration. Collectively, the noncoding loci converge on a set of core pathways, permitting a classification of tumors into pathway-based subtypes. The somatic eQTL network is disrupted in 88% of tumors, suggesting widespread impact of noncoding mutations in cancer.Analysis of whole-genome sequences and transcription data from tumors identifies noncoding loci in which mutations affect target gene expression. These somatic eQTLs can classify tumors into pathway-based subtypes and are disrupted in 88% of tumors.


Molecular Cancer Therapeutics | 2018

Disruption of NSD1 in head and neck cancer promotes favorable chemotherapeutic responses linked to hypomethylation

Nam Bui; Justin K. Huang; Ana Bojorquez-Gomez; Katherine Licon; Kyle Salinas Sanchez; Sean N. Tang; Alex N. Beckett; Tina Wang; Wei Zhang; John Paul Shen; Jason F. Kreisberg; Trey Ideker

Human papillomavirus (HPV)–negative head and neck squamous cell carcinoma (HNSCC) represents a distinct classification of cancer with worse expected outcomes. Of the 11 genes recurrently mutated in HNSCC, we identify a singular and substantial survival advantage for mutations in the gene encoding Nuclear Set Domain Containing Protein 1 (NSD1), a histone methyltransferase altered in approximately 10% of patients. This effect, a 55% decrease in risk of death in NSD1-mutated versus non-mutated patients, can be validated in an independent cohort. NSD1 alterations are strongly associated with widespread genome hypomethylation in the same tumors, to a degree not observed for any other mutated gene. To address whether NSD1 plays a causal role in these associations, we use CRISPR-Cas9 to disrupt NSD1 in HNSCC cell lines and find that this leads to substantial CpG hypomethylation and sensitivity to cisplatin, a standard chemotherapy in head and neck cancer, with a 40% to 50% decrease in the IC50 value. Such results are reinforced by a survey of 1,001 cancer cell lines, in which loss-of-function NSD1 mutations have an average 23% decrease in cisplatin IC50 value compared with cell lines with wild-type NSD1. Significance: This study identifies a favorable subtype of HPV–negative HNSCC linked to NSD1 mutation, hypomethylation, and cisplatin sensitivity. Mol Cancer Ther; 17(7); 1585–94. ©2018 AACR.

Collaboration


Dive into the John Paul Shen's collaboration.

Top Co-Authors

Avatar

Trey Ideker

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Matan Hofree

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jianfeng Li

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar

Prashant Mali

University of California

View shared research outputs
Researchain Logo
Decentralizing Knowledge