Network


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

Hotspot


Dive into the research topics where Eliezer M. Van Allen is active.

Publication


Featured researches published by Eliezer M. Van Allen.


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.


Science | 2015

Genomic correlates of response to CTLA-4 blockade in metastatic melanoma

Eliezer M. Van Allen; Diana Miao; Bastian Schilling; Sachet A. Shukla; Christian U. Blank; Lisa Zimmer; Antje Sucker; Uwe Hillen; Marnix H. Geukes Foppen; Simone M. Goldinger; Jochen Utikal; Jessica C. Hassel; Benjamin Weide; Katharina C. Kaehler; Carmen Loquai; Peter Mohr; Ralf Gutzmer; Reinhard Dummer; Stacey Gabriel; Catherine J. Wu; Dirk Schadendorf; Levi A. Garraway

Is cancer immunotherapy a private affair? Immune checkpoint blockade, a relatively new cancer treatment, substantially extends the survival of a subset of patients. Previous work has shown that patients whose tumors harbor the largest number of mutations—and thus produce a large number of “neoantigens” recognized as foreign by the immune system—are most likely to benefit. Expanding on these earlier studies, Van Allen et al. studied over 100 patients with melanoma and found a similar correlation (see the Perspective by Gubin and Schreiber). There was no evidence, however, that specific neoantigen sequences were shared by patients who responded. Science, this issue p. 207, see also p. 158 Melanoma patients who respond to immunotherapy do not appear to share common tumor neoantigens. [Also see Perspective by Gubin and Schreiber ] Monoclonal antibodies directed against cytotoxic T lymphocyte–associated antigen-4 (CTLA-4), such as ipilimumab, yield considerable clinical benefit for patients with metastatic melanoma by inhibiting immune checkpoint activity, but clinical predictors of response to these therapies remain incompletely characterized. To investigate the roles of tumor-specific neoantigens and alterations in the tumor microenvironment in the response to ipilimumab, we analyzed whole exomes from pretreatment melanoma tumor biopsies and matching germline tissue samples from 110 patients. For 40 of these patients, we also obtained and analyzed transcriptome data from the pretreatment tumor samples. Overall mutational load, neoantigen load, and expression of cytolytic markers in the immune microenvironment were significantly associated with clinical benefit. However, no recurrent neoantigen peptide sequences predicted responder patient populations. Thus, detailed integrated molecular characterization of large patient cohorts may be needed to identify robust determinants of response and resistance to immune checkpoint inhibitors.


Science | 2016

Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade

Nicholas McGranahan; Andrew Furness; Rachel Rosenthal; Sofie Ramskov; Rikke Birgitte Lyngaa; Sunil Kumar Saini; Mariam Jamal-Hanjani; Gareth A. Wilson; Nicolai Juul Birkbak; Crispin Hiley; Thomas B.K. Watkins; Seema Shafi; Nirupa Murugaesu; Richard Mitter; Ayse U. Akarca; Joseph Linares; Teresa Marafioti; Jake Y. Henry; Eliezer M. Van Allen; Diana Miao; Bastian Schilling; Dirk Schadendorf; Levi A. Garraway; Vladimir Makarov; Naiyer A. Rizvi; Alexandra Snyder; Matthew D. Hellmann; Taha Merghoub; Jedd D. Wolchok; Sachet A. Shukla

The cellular ancestry of tumor antigens One contributing factor in antitumor immunity is the repertoire of neoantigens created by genetic mutations within tumor cells. Like the corresponding mutations, these neoantigens show intratumoral heterogeneity. Some are present in all tumor cells (clonal), and others are present in only a fraction of cells (subclonal). In a study of lung cancer and melanoma, McGranahan et al. found that a high burden of clonal tumor neoantigens correlated with improved patient survival, an increased presence of tumor-infiltrating lymphocytes, and a durable response to immunotherapy. Science, this issue p. 1463 Analysis of the cellular ancestry of tumor neoantigens can predict which are most likely to induce an immune response. As tumors grow, they acquire mutations, some of which create neoantigens that influence the response of patients to immune checkpoint inhibitors. We explored the impact of neoantigen intratumor heterogeneity (ITH) on antitumor immunity. Through integrated analysis of ITH and neoantigen burden, we demonstrate a relationship between clonal neoantigen burden and overall survival in primary lung adenocarcinomas. CD8+ tumor-infiltrating lymphocytes reactive to clonal neoantigens were identified in early-stage non–small cell lung cancer and expressed high levels of PD-1. Sensitivity to PD-1 and CTLA-4 blockade in patients with advanced NSCLC and melanoma was enhanced in tumors enriched for clonal neoantigens. T cells recognizing clonal neoantigens were detectable in patients with durable clinical benefit. Cytotoxic chemotherapy–induced subclonal neoantigens, contributing to an increased mutational load, were enriched in certain poor responders. These data suggest that neoantigen heterogeneity may influence immune surveillance and support therapeutic developments targeting clonal neoantigens.


Science | 2016

Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-seq

Itay Tirosh; Benjamin Izar; Sanjay Prakadan; Marc H. Wadsworth; Daniel J. Treacy; John J. Trombetta; Asaf Rotem; Christopher Rodman; Christine G. Lian; George F. Murphy; Mohammad Fallahi-Sichani; Ken Dutton-Regester; Jia-Ren Lin; Ofir Cohen; Parin Shah; Diana Lu; Alex S. Genshaft; Travis K. Hughes; Carly G.K. Ziegler; Samuel W. Kazer; Aleth Gaillard; Kellie E. Kolb; Alexandra-Chloé Villani; Cory M. Johannessen; Aleksandr Andreev; Eliezer M. Van Allen; Monica M. Bertagnolli; Peter K. Sorger; Ryan J. Sullivan; Keith T. Flaherty

Single-cell expression profiles of melanoma Tumors harbor multiple cell types that are thought to play a role in the development of resistance to drug treatments. Tirosh et al. used single-cell sequencing to investigate the distribution of these differing genetic profiles within melanomas. Many cells harbored heterogeneous genetic programs that reflected two different states of genetic expression, one of which was linked to resistance development. Following drug treatment, the resistance-linked expression state was found at a much higher level. Furthermore, the environment of the melanoma cells affected their gene expression programs. Science, this issue p. 189 Melanoma cells show transcriptional heterogeneity. To explore the distinct genotypic and phenotypic states of melanoma tumors, we applied single-cell RNA sequencing (RNA-seq) to 4645 single cells isolated from 19 patients, profiling malignant, immune, stromal, and endothelial cells. Malignant cells within the same tumor displayed transcriptional heterogeneity associated with the cell cycle, spatial context, and a drug-resistance program. In particular, all tumors harbored malignant cells from two distinct transcriptional cell states, such that tumors characterized by high levels of the MITF transcription factor also contained cells with low MITF and elevated levels of the AXL kinase. Single-cell analyses suggested distinct tumor microenvironmental patterns, including cell-to-cell interactions. Analysis of tumor-infiltrating T cells revealed exhaustion programs, their connection to T cell activation and clonal expansion, and their variability across patients. Overall, we begin to unravel the cellular ecosystem of tumors and how single-cell genomics offers insights with implications for both targeted and immune therapies.


Nature Biotechnology | 2014

Whole exome sequencing of circulating tumor cells provides a window into metastatic prostate cancer

Jens Lohr; Viktor A. Adalsteinsson; Kristian Cibulskis; Atish D. Choudhury; Mara Rosenberg; Peter Cruz-Gordillo; Joshua M. Francis; Cheng-Zhong Zhang; Alex K. Shalek; Rahul Satija; John J. Trombetta; Diana Lu; Naren Tallapragada; Narmin Tahirova; Sora Kim; Brendan Blumenstiel; Carrie Sougnez; Alarice Lowe; Bang Wong; Daniel Auclair; Eliezer M. Van Allen; Mari Nakabayashi; Rosina T. Lis; Gwo-Shu Mary Lee; Tiantian Li; Matthew S. Chabot; Amy Ly; Mary-Ellen Taplin; Thomas E. Clancy; Massimo Loda

Comprehensive analyses of cancer genomes promise to inform prognoses and precise cancer treatments. A major barrier, however, is inaccessibility of metastatic tissue. A potential solution is to characterize circulating tumor cells (CTCs), but this requires overcoming the challenges of isolating rare cells and sequencing low-input material. Here we report an integrated process to isolate, qualify and sequence whole exomes of CTCs with high fidelity using a census-based sequencing strategy. Power calculations suggest that mapping of >99.995% of the standard exome is possible in CTCs. We validated our process in two patients with prostate cancer, including one for whom we sequenced CTCs, a lymph node metastasis and nine cores of the primary tumor. Fifty-one of 73 CTC mutations (70%) were present in matched tissue. Moreover, we identified 10 early trunk and 56 metastatic trunk mutations in the non-CTC tumor samples and found 90% and 73% of these mutations, respectively, in CTC exomes. This study establishes a foundation for CTC genomics in the clinic.


Cancer Discovery | 2014

MAP Kinase Pathway Alterations in BRAF-Mutant Melanoma Patients with Acquired Resistance to Combined RAF/MEK Inhibition

Nikhil Wagle; Eliezer M. Van Allen; Daniel J. Treacy; Dennie T. Frederick; Zachary A. Cooper; Amaro Taylor-Weiner; Mara Rosenberg; Eva M. Goetz; Ryan J. Sullivan; Deborah N. Farlow; Dennis Friedrich; Kristin Anderka; Danielle Perrin; Cory M. Johannessen; Aaron McKenna; Kristian Cibulskis; Gregory V. Kryukov; Eran Hodis; Donald P. Lawrence; Sheila Fisher; Gad Getz; Stacey Gabriel; Scott L. Carter; Keith T. Flaherty; Jennifer A. Wargo; Levi A. Garraway

Treatment of BRAF-mutant melanoma with combined dabrafenib and trametinib, which target RAF and the downstream MAP-ERK kinase (MEK)1 and MEK2 kinases, respectively, improves progression-free survival and response rates compared with dabrafenib monotherapy. Mechanisms of clinical resistance to combined RAF/MEK inhibition are unknown. We performed whole-exome sequencing (WES) and whole-transcriptome sequencing (RNA-seq) on pretreatment and drug-resistant tumors from five patients with acquired resistance to dabrafenib/trametinib. In three of these patients, we identified additional mitogen-activated protein kinase (MAPK) pathway alterations in the resistant tumor that were not detected in the pretreatment tumor, including a novel activating mutation in MEK2 (MEK2(Q60P)). MEK2(Q60P) conferred resistance to combined RAF/MEK inhibition in vitro, but remained sensitive to inhibition of the downstream kinase extracellular signal-regulated kinase (ERK). The continued MAPK signaling-based resistance identified in these patients suggests that alternative dosing of current agents, more potent RAF/MEK inhibitors, and/or inhibition of the downstream kinase ERK may be needed for durable control of BRAF-mutant melanoma.


Cancer Discovery | 2015

Genomic Characterization of Brain Metastases Reveals Branched Evolution and Potential Therapeutic Targets

Priscilla K. Brastianos; Scott L. Carter; Sandro Santagata; Daniel P. Cahill; Amaro Taylor-Weiner; Robert T. Jones; Eliezer M. Van Allen; Michael S. Lawrence; Peleg Horowitz; Kristian Cibulskis; Keith L. Ligon; Josep Tabernero; Joan Seoane; Elena Martinez-Saez; William T. Curry; Ian F. Dunn; Sun Ha Paek; Sung-Hye Park; Aaron McKenna; Aaron Chevalier; Mara Rosenberg; Fred G. Barker; Corey M. Gill; Paul Van Hummelen; Aaron R. Thorner; Bruce E. Johnson; Mai P. Hoang; Toni K. Choueiri; Sabina Signoretti; Carrie Sougnez

UNLABELLED Brain metastases are associated with a dismal prognosis. Whether brain metastases harbor distinct genetic alterations beyond those observed in primary tumors is unknown. We performed whole-exome sequencing of 86 matched brain metastases, primary tumors, and normal tissue. In all clonally related cancer samples, we observed branched evolution, where all metastatic and primary sites shared a common ancestor yet continued to evolve independently. In 53% of cases, we found potentially clinically informative alterations in the brain metastases not detected in the matched primary-tumor sample. In contrast, spatially and temporally separated brain metastasis sites were genetically homogenous. Distal extracranial and regional lymph node metastases were highly divergent from brain metastases. We detected alterations associated with sensitivity to PI3K/AKT/mTOR, CDK, and HER2/EGFR inhibitors in the brain metastases. Genomic analysis of brain metastases provides an opportunity to identify potentially clinically informative alterations not detected in clinically sampled primary tumors, regional lymph nodes, or extracranial metastases. SIGNIFICANCE Decisions for individualized therapies in patients with brain metastasis are often made from primary-tumor biopsies. We demonstrate that clinically actionable alterations present in brain metastases are frequently not detected in primary biopsies, suggesting that sequencing of primary biopsies alone may miss a substantial number of opportunities for targeted therapy.


Cancer Discovery | 2013

A Genome-Scale RNA Interference Screen Implicates NF1 Loss in Resistance to RAF Inhibition

Steven Whittaker; Jean-Philippe Theurillat; Eliezer M. Van Allen; Nikhil Wagle; Jessica Hsiao; Glenn S. Cowley; Dirk Schadendorf; David E. Root; Levi A. Garraway

RAF inhibitors such as vemurafenib and dabrafenib block BRAF-mediated cell proliferation and achieve meaningful clinical benefit in the vast majority of patients with BRAF(V600E)-mutant melanoma. However, some patients do not respond to this regimen, and nearly all progress to therapeutic resistance. We used a pooled RNA interference screen targeting more than 16,500 genes to discover loss-of-function events that could drive resistance to RAF inhibition. The highest ranking gene was NF1, which encodes neurofibromin, a tumor suppressor that inhibits RAS activity. NF1 loss mediates resistance to RAF and mitogen-activated protein kinase (MAPK) kinase kinase (MEK) inhibitors through sustained MAPK pathway activation. However, cells lacking NF1 retained sensitivity to the irreversible RAF inhibitor AZ628 and an ERK inhibitor. NF1 mutations were observed in BRAF-mutant tumor cells that are intrinsically resistant to RAF inhibition and in melanoma tumors obtained from patients exhibiting resistance to vemurafenib, thus showing the clinical potential for NF1-driven resistance to RAF/MEK-targeted therapies.


The New England Journal of Medicine | 2014

Response and Acquired Resistance to Everolimus in Anaplastic Thyroid Cancer

Nikhil Wagle; Brian C. Grabiner; Eliezer M. Van Allen; Ali Amin-Mansour; Amaro Taylor-Weiner; Mara Rosenberg; Nathanael S. Gray; Justine A. Barletta; Yanan Guo; Scott J. Swanson; Daniel T. Ruan; Glenn J. Hanna; Robert I. Haddad; Gad Getz; David J. Kwiatkowski; Scott L. Carter; David M. Sabatini; Pasi A. Jänne; Levi A. Garraway; Jochen H. Lorch

Everolimus, an inhibitor of the mammalian target of rapamycin (mTOR), is effective in treating tumors harboring alterations in the mTOR pathway. Mechanisms of resistance to everolimus remain undefined. Resistance developed in a patient with metastatic anaplastic thyroid carcinoma after an extraordinary 18-month response. Whole-exome sequencing of pretreatment and drug-resistant tumors revealed a nonsense mutation in TSC2, a negative regulator of mTOR, suggesting a mechanism for exquisite sensitivity to everolimus. The resistant tumor also harbored a mutation in MTOR that confers resistance to allosteric mTOR inhibition. The mutation remains sensitive to mTOR kinase inhibitors.


Nature Genetics | 2014

RNF43 is frequently mutated in colorectal and endometrial cancers

Marios Giannakis; Eran Hodis; Xinmeng Jasmine Mu; Mai Yamauchi; Joseph Rosenbluh; Kristian Cibulskis; Gordon Saksena; Michael S. Lawrence; Zhi Rong Qian; Reiko Nishihara; Eliezer M. Van Allen; William C. Hahn; Stacey Gabriel; Eric S. Lander; Gad Getz; Shuji Ogino; Charles S. Fuchs; Levi A. Garraway

We report somatic mutations of RNF43 in over 18% of colorectal adenocarcinomas and endometrial carcinomas. RNF43 encodes an E3 ubiquitin ligase that negatively regulates Wnt signaling. Truncating mutations of RNF43 are more prevalent in microsatellite-unstable tumors and show mutual exclusivity with inactivating APC mutations in colorectal adenocarcinomas. These results indicate that RNF43 is one of the most commonly mutated genes in colorectal and endometrial cancers.

Collaboration


Dive into the Eliezer M. Van Allen's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jonathan E. Rosenberg

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge