Network


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

Hotspot


Dive into the research topics where John T. Poirier is active.

Publication


Featured researches published by John T. Poirier.


Nature Genetics | 2012

Comprehensive genomic analysis identifies SOX2 as a frequently amplified gene in small-cell lung cancer

Charles M. Rudin; Steffen Durinck; Eric Stawiski; John T. Poirier; Zora Modrusan; David S. Shames; Emily Bergbower; Yinghui Guan; James Shin; Joseph Guillory; Celina Sanchez Rivers; Catherine K. Foo; Deepali Bhatt; Jeremy Stinson; Florian Gnad; Peter M. Haverty; Robert Gentleman; Subhra Chaudhuri; Vasantharajan Janakiraman; Bijay S. Jaiswal; Chaitali Parikh; Wenlin Yuan; Zemin Zhang; Hartmut Koeppen; Thomas D. Wu; Howard M. Stern; Robert L. Yauch; Kenneth Huffman; Diego D Paskulin; Peter B. Illei

Small-cell lung cancer (SCLC) is an exceptionally aggressive disease with poor prognosis. Here, we obtained exome, transcriptome and copy-number alteration data from approximately 53 samples consisting of 36 primary human SCLC and normal tissue pairs and 17 matched SCLC and lymphoblastoid cell lines. We also obtained data for 4 primary tumors and 23 SCLC cell lines. We identified 22 significantly mutated genes in SCLC, including genes encoding kinases, G protein–coupled receptors and chromatin-modifying proteins. We found that several members of the SOX family of genes were mutated in SCLC. We also found SOX2 amplification in ∼27% of the samples. Suppression of SOX2 using shRNAs blocked proliferation of SOX2-amplified SCLC lines. RNA sequencing identified multiple fusion transcripts and a recurrent RLF-MYCL1 fusion. Silencing of MYCL1 in SCLC cell lines that had the RLF-MYCL1 fusion decreased cell proliferation. These data provide an in-depth view of the spectrum of genomic alterations in SCLC and identify several potential targets for therapeutic intervention.


Nature Communications | 2015

RB loss in resistant EGFR mutant lung adenocarcinomas that transform to small-cell lung cancer

Matthew J. Niederst; Lecia V. Sequist; John T. Poirier; Craig H. Mermel; Elizabeth L. Lockerman; Angel R. Garcia; Ryohei Katayama; Carlotta Costa; Kenneth N. Ross; Teresa Moran; Emily Howe; L. Fulton; Hillary Mulvey; Lindsay A. Bernardo; Farhiya Mohamoud; Norikatsu Miyoshi; Paul A. VanderLaan; Daniel B. Costa; Pasi A. Jänne; Darrell R. Borger; Sridhar Ramaswamy; Toshi Shioda; Anthony John Iafrate; Gad Getz; Charles M. Rudin; Mari Mino-Kenudson; Jeffrey A. Engelman

Tyrosine kinase inhibitors are effective treatments for non-small-cell lung cancers (NSCLCs) with epidermal growth factor receptor (EGFR) mutations. However, relapse typically occurs after an average of 1 year of continuous treatment. A fundamental histological transformation from NSCLC to small-cell lung cancer (SCLC) is observed in a subset of the resistant cancers, but the molecular changes associated with this transformation remain unknown. Analysis of tumour samples and cell lines derived from resistant EGFR mutant patients revealed that Retinoblastoma (RB) is lost in 100% of these SCLC transformed cases, but rarely in those that remain NSCLC. Further, increased neuroendocrine marker and decreased EGFR expression as well as greater sensitivity to BCL2 family inhibition are observed in resistant SCLC transformed cancers compared with resistant NSCLCs. Together, these findings suggest that this subset of resistant cancers ultimately adopt many of the molecular and phenotypic characteristics of classical SCLC.


Nature | 2016

A combinatorial strategy for treating KRAS-mutant lung cancer

Eusebio Manchado; Susann Weissmueller; John P. Morris; Chi Chao Chen; Ramona Wullenkord; Amaia Lujambio; Elisa de Stanchina; John T. Poirier; Justin F. Gainor; Ryan B. Corcoran; Jeffrey A. Engelman; Charles M. Rudin; Neal Rosen; Scott W. Lowe

Therapeutic targeting of KRAS-mutant lung adenocarcinoma represents a major goal of clinical oncology. KRAS itself has proved difficult to inhibit, and the effectiveness of agents that target key KRAS effectors has been thwarted by activation of compensatory or parallel pathways that limit their efficacy as single agents. Here we take a systematic approach towards identifying combination targets for trametinib, a MEK inhibitor approved by the US Food and Drug Administration, which acts downstream of KRAS to suppress signalling through the mitogen-activated protein kinase (MAPK) cascade. Informed by a short-hairpin RNA screen, we show that trametinib provokes a compensatory response involving the fibroblast growth factor receptor 1 (FGFR1) that leads to signalling rebound and adaptive drug resistance. As a consequence, genetic or pharmacological inhibition of FGFR1 in combination with trametinib enhances tumour cell death in vitro and in vivo. This compensatory response shows distinct specificities: it is dominated by FGFR1 in KRAS-mutant lung and pancreatic cancer cells, but is not activated or involves other mechanisms in KRAS wild-type lung and KRAS-mutant colon cancer cells. Importantly, KRAS-mutant lung cancer cells and patients’ tumours treated with trametinib show an increase in FRS2 phosphorylation, a biomarker of FGFR activation; this increase is abolished by FGFR1 inhibition and correlates with sensitivity to trametinib and FGFR inhibitor combinations. These results demonstrate that FGFR1 can mediate adaptive resistance to trametinib and validate a combinatorial approach for treating KRAS-mutant lung cancer.


Clinical Cancer Research | 2016

Next-Generation Sequencing of Pulmonary Large Cell Neuroendocrine Carcinoma Reveals Small Cell Carcinoma–like and Non–Small Cell Carcinoma–like Subsets

Natasha Rekhtman; Maria Catherine Pietanza; Matthew D. Hellmann; Jarushka Naidoo; Arshi Arora; Helen H. Won; Darragh Halpenny; Hangjun Wang; Shaozhou K. Tian; Anya Litvak; Paul K. Paik; Alexander Drilon; Nicholas D. Socci; John T. Poirier; Ronglai Shen; Michael F. Berger; Andre L. Moreira; William D. Travis; Charles M. Rudin; Marc Ladanyi

Purpose: Pulmonary large cell neuroendocrine carcinoma (LCNEC) is a highly aggressive neoplasm, whose biologic relationship to small cell lung carcinoma (SCLC) versus non-SCLC (NSCLC) remains unclear, contributing to uncertainty regarding optimal clinical management. To clarify these relationships, we analyzed genomic alterations in LCNEC compared with other major lung carcinoma types. Experimental Design: LCNEC (n = 45) tumor/normal pairs underwent targeted next-generation sequencing of 241 cancer genes by Memorial Sloan Kettering-Integrated Mutation Profiling of Actionable Cancer Targets (MSK-IMPACT) platform and comprehensive histologic, immunohistochemical, and clinical analysis. Genomic data were compared with MSK-IMPACT analysis of other lung carcinoma histologies (n = 242). Results: Commonly altered genes in LCNEC included TP53 (78%), RB1 (38%), STK11 (33%), KEAP1 (31%), and KRAS (22%). Genomic profiles segregated LCNEC into 2 major and 1 minor subsets: SCLC-like (n = 18), characterized by TP53+RB1 co-mutation/loss and other SCLC-type alterations, including MYCL amplification; NSCLC-like (n = 25), characterized by the lack of coaltered TP53+RB1 and nearly universal occurrence of NSCLC-type mutations (STK11, KRAS, and KEAP1); and carcinoid-like (n = 2), characterized by MEN1 mutations and low mutation burden. SCLC-like and NSCLC-like subsets revealed several clinicopathologic differences, including higher proliferative activity in SCLC-like tumors (P < 0.0001) and exclusive adenocarcinoma-type differentiation marker expression in NSCLC-like tumors (P = 0.005). While exhibiting predominant similarity with lung adenocarcinoma, NSCLC-like LCNEC harbored several distinctive genomic alterations, including more frequent mutations in NOTCH family genes (28%), implicated as key regulators of neuroendocrine differentiation. Conclusions: LCNEC is a biologically heterogeneous group of tumors, comprising distinct subsets with genomic signatures of SCLC, NSCLC (predominantly adenocarcinoma), and rarely, highly proliferative carcinoids. Recognition of these subsets may inform the classification and management of LCNEC patients. Clin Cancer Res; 22(14); 3618–29. ©2016 AACR.


Clinical Cancer Research | 2011

Phase I Clinical Study of Seneca Valley Virus (SVV-001), a Replication-Competent Picornavirus, in Advanced Solid Tumors with Neuroendocrine Features

Charles M. Rudin; John T. Poirier; Neil Senzer; J. Stephenson; David M. Loesch; Kevin D. Burroughs; P. Seshidhar Reddy; Christine L. Hann; Paul L. Hallenbeck

Purpose: Seneca Valley Virus (SVV-001) is a novel naturally occurring replication-competent picornavirus with potent and selective tropism for neuroendocrine cancer cell types, including small cell lung cancer. We conducted a first-in-human, first-in-class phase I clinical trial of this agent in patients with cancers with neuroendocrine features, including small cell lung cancer. Experimental Design: Clinical evaluation of single intravenous doses in patients with cancers with neuroendocrine features was performed across five log-increments from 107 to 1011 vp/kg. Toxicity, viral titers and clearance, neutralizing antibody development, and tumor response were assessed. Results: A total of 30 patients were treated with SVV-001, including six with small cell carcinoma at the lowest dose of 107 vp/kg. SVV-001 was well tolerated, with no dose-limiting toxicities observed in any dose cohort. Viral clearance was documented in all subjects and correlated temporally with development of antiviral antibodies. Evidence of in vivo intratumoral viral replication was observed among patients with small cell carcinoma, with peak viral titers estimated to be >103-fold higher than the administered dose. One patient with previously progressive chemorefractory small cell lung cancer remained progression-free for 10 months after SVV-001 administration, and is alive over 3 years after treatment. Conclusions: Intravenous SVV-001 administration in patients is well tolerated at doses up to 1011 vp/kg, with predictable viral clearance kinetics, intratumoral viral replication, and evidence of antitumor activity in patients with small cell lung cancer. Phase II clinical evaluation in small cell lung cancer is warranted, and has been initiated. Clin Cancer Res; 17(4); 888–95. ©2011 AACR.


Oncogene | 2015

DNA methylation in small cell lung cancer defines distinct disease subtypes and correlates with high expression of EZH2

John T. Poirier; Eric E. Gardner; N Connis; A L Moreira; E. de Stanchina; C L Hann; Charles M. Rudin

Small cell lung cancer (SCLC) is an aggressive malignancy characterized by early metastasis, rapid development of resistance to chemotherapy and genetic instability. This study profiles DNA methylation in SCLC, patient-derived xenografts (PDX) and cell lines at single-nucleotide resolution. DNA methylation patterns of primary samples are distinct from those of cell lines, whereas PDX maintain a pattern closely consistent with primary samples. Clustering of DNA methylation and gene expression of primary SCLC revealed distinct disease subtypes among histologically indistinguishable primary patient samples with similar genetic alterations. SCLC is notable for dense clustering of high-level methylation in discrete promoter CpG islands, in a pattern clearly distinct from other lung cancers and strongly correlated with high expression of the E2F target and histone methyltransferase gene EZH2. Pharmacologic inhibition of EZH2 in a SCLC PDX markedly inhibited tumor growth.


Molecular Cancer Research | 2009

Upregulation of MMP-2 by HMGA1 Promotes Transformation in Undifferentiated, Large-Cell Lung Cancer

Joelle Hillion; Lisa Wood; Mita Mukherjee; Raka Bhattacharya; Francescopaolo Di Cello; Jeanne Kowalski; Ossama Elbahloul; Jodi B. Segal; John T. Poirier; Charles M. Rudin; Surajit Dhara; Amy Belton; Biju Joseph; Stanley Zucker; Linda M. S. Resar

Although lung cancer is the leading cause of cancer death worldwide, the precise molecular mechanisms that give rise to lung cancer are incompletely understood. Here, we show that HMGA1 is an important oncogene that drives transformation in undifferentiated, large-cell carcinoma. First, we show that the HMGA1 gene is overexpressed in lung cancer cell lines and primary human lung tumors. Forced overexpression of HMGA1 induces a transformed phenotype with anchorage-independent cell growth in cultured lung cells derived from normal tissue. Conversely, inhibiting HMGA1 expression blocks anchorage-independent cell growth in the H1299 metastatic, undifferentiated, large-cell human lung carcinoma cells. We also show that the matrix metalloproteinase-2 (MMP-2) gene is a downstream target upregulated by HMGA1 in large-cell carcinoma cells. In chromatin immunoprecipitation experiments, HMGA1 binds directly to the MMP-2 promoter in vivo in large-cell lung cancer cells, but not in squamous cell carcinoma cells. In large-cell carcinoma cell lines, there is a significant, positive correlation between HMGA1 and MMP-2 mRNA. Moreover, interfering with MMP-2 expression blocks anchorage-independent cell growth in H1299 large-cell carcinoma cells, indicating that the HMGA1–MMP-2 pathway is required for this transformation phenotype in these cells. Blocking MMP-2 expression also inhibits migration and invasion in the H1299 large-cell carcinoma cells. Our findings suggest an important role for MMP-2 in transformation mediated by HMGA1 in large-cell, undifferentiated lung carcinoma and support the development of strategies to target this pathway in selected tumors. (Mol Cancer Res 2009;7(11):1803–12)


Journal of Clinical Investigation | 2016

CD47-blocking immunotherapies stimulate macrophage-mediated destruction of small-cell lung cancer.

Kipp Weiskopf; Nadine S. Jahchan; Peter J. Schnorr; Sandra Cristea; Aaron M. Ring; Roy L. Maute; Anne K. Volkmer; Jens Peter Volkmer; Jie Liu; Jing Shan Lim; Dian Yang; Garrett Seitz; Thuyen Nguyen; Di Wu; Kevin M. Jude; Heather Guerston; Francesca Trapani; Julie George; John T. Poirier; Eric E. Gardner; Linde A. Miles; Elisa de Stanchina; Shane Lofgren; Hannes Vogel; Monte M. Winslow; Caroline Dive; Roman K. Thomas; Charles M. Rudin; Matt Van De Rijn; Ravindra Majeti

Small-cell lung cancer (SCLC) is a highly aggressive subtype of lung cancer with limited treatment options. CD47 is a cell-surface molecule that promotes immune evasion by engaging signal-regulatory protein alpha (SIRPα), which serves as an inhibitory receptor on macrophages. Here, we found that CD47 is highly expressed on the surface of human SCLC cells; therefore, we investigated CD47-blocking immunotherapies as a potential approach for SCLC treatment. Disruption of the interaction of CD47 with SIRPα using anti-CD47 antibodies induced macrophage-mediated phagocytosis of human SCLC patient cells in culture. In a murine model, administration of CD47-blocking antibodies or targeted inactivation of the Cd47 gene markedly inhibited SCLC tumor growth. Furthermore, using comprehensive antibody arrays, we identified several possible therapeutic targets on the surface of SCLC cells. Antibodies to these targets, including CD56/neural cell adhesion molecule (NCAM), promoted phagocytosis in human SCLC cell lines that was enhanced when combined with CD47-blocking therapies. In light of recent clinical trials for CD47-blocking therapies in cancer treatment, these findings identify disruption of the CD47/SIRPα axis as a potential immunotherapeutic strategy for SCLC. This approach could enable personalized immunotherapeutic regimens in patients with SCLC and other cancers.


Journal of Thoracic Oncology | 2016

Small Cell Lung Cancer: Can Recent Advances in Biology and Molecular Biology Be Translated into Improved Outcomes?

Paul A. Bunn; John D. Minna; Alexander Augustyn; Adi F. Gazdar; Youcef Ouadah; Mark A. Krasnow; Anton Berns; Elisabeth Brambilla; Natasha Rekhtman; Pierre P. Massion; Matthew J. Niederst; Martin Peifer; Jun Yokota; Ramaswamy Govindan; John T. Poirier; Lauren Averett Byers; Murry W. Wynes; David G. McFadden; David MacPherson; Christine L. Hann; Anna F. Farago; Caroline Dive; Beverly A. Teicher; Craig D. Peacock; Jane E. Johnson; Melanie H. Cobb; Hans Guido Wendel; David R. Spigel; Julien Sage; Ping Yang

Paul A. Bunn Jr., MD, John D. Minna, MD, Alexander Augustyn, PhD, Adi F. Gazdar, MD, Youcef Ouadah, BS, Mark A. Krasnow, MD, PhD, Anton Berns, PhD, Elisabeth Brambilla, MD, Natasha Rekhtman, MD, PhD, Pierre P. Massion, MD, Matthew Niederst, PhD, Martin Peifer, PhD, Jun Yokota, MD, Ramaswamy Govindan, MD, John T. Poirier, PhD, Lauren A. Byers, MD, Murry W. Wynes, PhD, David G. McFadden, MD, PhD, David MacPherson, PhD, Christine L. Hann, MD, PhD, Anna F. Farago, MD, PhD, Caroline Dive, PhD, Beverly A. Teicher, PhD, Craig D. Peacock, PhD, Jane E. Johnson, PhD, Melanie H. Cobb, PhD, Hans-Guido Wendel, MD, David Spigel, MD, Julien Sage, PhD, Ping Yang, MD, PhD, M. Catherine Pietanza, MD, Lee M. Krug, MD, John Heymach, MD, PhD, Peter Ujhazy, MD, PhD, Caicun Zhou, MD, PhD, Koichi Goto, MD, Afshin Dowlati, MD, Camilla Laulund Christensen, PhD, Keunchil Park, MD, PhD, Lawrence H. Einhorn, MD, Martin J. Edelman, MD, Giuseppe Giaccone, MD, PhD, David E. Gerber, MD, Ravi Salgia, MD, PhD, Taofeek Owonikoko, MD, PhD, Shakun Malik, MD, Niki Karachaliou, MD, David R. Gandara, MD, Ben J. Slotman, MD, PhD, Fiona Blackhall, MD, PhD, Glenwood Goss, MD, FRCPC, Roman Thomas, MD, Charles M. Rudin, MD, PhD, Fred R. Hirsch, MD, PhD*


Nature Medicine | 2017

Keap1 loss promotes Kras -driven lung cancer and results in dependence on glutaminolysis

Rodrigo Romero; Volkan I. Sayin; Shawn M. Davidson; Matthew R. Bauer; Simranjit X. Singh; Sarah LeBoeuf; Triantafyllia R. Karakousi; Donald Christian Ellis; Arjun Bhutkar; Francisco J. Sánchez-Rivera; Lakshmipriya Subbaraj; Britney Martinez; Roderick T. Bronson; Justin R. Prigge; Edward E. Schmidt; Craig J. Thomas; Chandra Goparaju; Angela M. Davies; Igor Dolgalev; Adriana Heguy; Viola Allaj; John T. Poirier; Andre L. Moreira; Charles M. Rudin; Harvey I. Pass; Matthew G. Vander Heiden; Tyler Jacks; Thales Papagiannakopoulos

Treating KRAS-mutant lung adenocarcinoma (LUAD) remains a major challenge in cancer treatment given the difficulties associated with directly inhibiting the KRAS oncoprotein. One approach to addressing this challenge is to define mutations that frequently co-occur with those in KRAS, which themselves may lead to therapeutic vulnerabilities in tumors. Approximately 20% of KRAS-mutant LUAD tumors carry loss-of-function mutations in the KEAP1 gene encoding Kelch-like ECH-associated protein 1 (refs. 2, 3, 4), a negative regulator of nuclear factor erythroid 2-like 2 (NFE2L2; hereafter NRF2), which is the master transcriptional regulator of the endogenous antioxidant response. The high frequency of mutations in KEAP1 suggests an important role for the oxidative stress response in lung tumorigenesis. Using a CRISPR–Cas9-based approach in a mouse model of KRAS-driven LUAD, we examined the effects of Keap1 loss in lung cancer progression. We show that loss of Keap1 hyperactivates NRF2 and promotes KRAS-driven LUAD in mice. Through a combination of CRISPR–Cas9-based genetic screening and metabolomic analyses, we show that Keap1- or Nrf2-mutant cancers are dependent on increased glutaminolysis, and this property can be therapeutically exploited through the pharmacological inhibition of glutaminase. Finally, we provide a rationale for stratification of human patients with lung cancer harboring KRAS/KEAP1- or KRAS/NRF2-mutant lung tumors as likely to respond to glutaminase inhibition.

Collaboration


Dive into the John T. Poirier's collaboration.

Top Co-Authors

Avatar

Charles M. Rudin

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Benjamin H. Lok

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Elisa de Stanchina

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Eric E. Gardner

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jason S. Lewis

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Linde A. Miles

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar

Natasha Rekhtman

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Viola Allaj

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge