Network


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

Hotspot


Dive into the research topics where Sasha Pantel is active.

Publication


Featured researches published by Sasha Pantel.


Cancer Discovery | 2016

Genomic Copy Number Dictates a Gene-Independent Cell Response to CRISPR/Cas9 Targeting

Andrew J. Aguirre; Robin M. Meyers; Barbara A. Weir; Francisca Vazquez; Cheng-Zhong Zhang; Uri Ben-David; April Cook; Gavin Ha; William F. Harrington; Mihir Doshi; Maria Kost-Alimova; Stanley Gill; Han Xu; Levi D. Ali; Guozhi Jiang; Sasha Pantel; Yenarae Lee; Amy Goodale; Andrew D. Cherniack; Coyin Oh; Gregory V. Kryukov; Glenn S. Cowley; Levi A. Garraway; Kimberly Stegmaier; Charles W. M. Roberts; Todd R. Golub; Matthew Meyerson; David E. Root; Aviad Tsherniak; William C. Hahn

UNLABELLED The CRISPR/Cas9 system enables genome editing and somatic cell genetic screens in mammalian cells. We performed genome-scale loss-of-function screens in 33 cancer cell lines to identify genes essential for proliferation/survival and found a strong correlation between increased gene copy number and decreased cell viability after genome editing. Within regions of copy-number gain, CRISPR/Cas9 targeting of both expressed and unexpressed genes, as well as intergenic loci, led to significantly decreased cell proliferation through induction of a G2 cell-cycle arrest. By examining single-guide RNAs that map to multiple genomic sites, we found that this cell response to CRISPR/Cas9 editing correlated strongly with the number of target loci. These observations indicate that genome targeting by CRISPR/Cas9 elicits a gene-independent antiproliferative cell response. This effect has important practical implications for the interpretation of CRISPR/Cas9 screening data and confounds the use of this technology for the identification of essential genes in amplified regions. SIGNIFICANCE We found that the number of CRISPR/Cas9-induced DNA breaks dictates a gene-independent antiproliferative response in cells. These observations have practical implications for using CRISPR/Cas9 to interrogate cancer gene function and illustrate that cancer cells are highly sensitive to site-specific DNA damage, which may provide a path to novel therapeutic strategies. Cancer Discov; 6(8); 914-29. ©2016 AACR.See related commentary by Sheel and Xue, p. 824See related article by Munoz et al., p. 900This article is highlighted in the In This Issue feature, p. 803.


Cancer Cell | 2016

High-throughput Phenotyping of Lung Cancer Somatic Mutations

Alice H. Berger; Angela N. Brooks; Xiaoyun Wu; Yashaswi Shrestha; Candace R. Chouinard; Federica Piccioni; Mukta Bagul; Atanas Kamburov; Marcin Imielinski; Larson Hogstrom; Cong Zhu; Xiaoping Yang; Sasha Pantel; Ryo Sakai; Jacqueline Watson; Nathan Kaplan; Joshua D. Campbell; Shantanu Singh; David E. Root; Rajiv Narayan; Ted Natoli; David L. Lahr; Itay Tirosh; Pablo Tamayo; Gad Getz; Bang Wong; John G. Doench; Aravind Subramanian; Todd R. Golub; Matthew Meyerson

Recent genome sequencing efforts have identified millions of somatic mutations in cancer. However, the functional impact of most variants is poorly understood. Here we characterize 194 somatic mutations identified in primary lung adenocarcinomas. We present an expression-based variant-impact phenotyping (eVIP) method that uses gene expression changes to distinguish impactful from neutral somatic mutations. eVIP identified 69% of mutations analyzed as impactful and 31% as functionally neutral. A subset of the impactful mutations induces xenograft tumor formation in mice and/or confers resistance to cellular EGFR inhibition. Among these impactful variants are rare somatic, clinically actionable variants including EGFR S645C, ARAF S214C and S214F, ERBB2 S418T, and multiple BRAF variants, demonstrating that rare mutations can be functionally important in cancer.


Nature Genetics | 2017

Computational correction of copy number effect improves specificity of CRISPR-Cas9 essentiality screens in cancer cells

Robin M. Meyers; Jordan Bryan; James M McFarland; Barbara A. Weir; Ann E. Sizemore; Han Xu; Neekesh V. Dharia; Phillip G Montgomery; Glenn S. Cowley; Sasha Pantel; Amy Goodale; Yenarae Lee; Levi D. Ali; Guozhi Jiang; Rakela Lubonja; William F. Harrington; Matthew R. Strickland; Ting Wu; Derek C Hawes; Victor A Zhivich; Meghan R Wyatt; Zohra Kalani; Jaime J Chang; Michael Okamoto; Kimberly Stegmaier; Todd R. Golub; Jesse S. Boehm; Francisca Vazquez; David E. Root; William C. Hahn

The CRISPR–Cas9 system has revolutionized gene editing both at single genes and in multiplexed loss-of-function screens, thus enabling precise genome-scale identification of genes essential for proliferation and survival of cancer cells. However, previous studies have reported that a gene-independent antiproliferative effect of Cas9-mediated DNA cleavage confounds such measurement of genetic dependency, thereby leading to false-positive results in copy number–amplified regions. We developed CERES, a computational method to estimate gene-dependency levels from CRISPR–Cas9 essentiality screens while accounting for the copy number–specific effect. In our efforts to define a cancer dependency map, we performed genome-scale CRISPR–Cas9 essentiality screens across 342 cancer cell lines and applied CERES to this data set. We found that CERES decreased false-positive results and estimated sgRNA activity for both this data set and previously published screens performed with different sgRNA libraries. We further demonstrate the utility of this collection of screens, after CERES correction, for identifying cancer-type-specific vulnerabilities.


Cell Reports | 2016

Phenotypic Characterization of a Comprehensive Set of MAPK1/ERK2 Missense Mutants

Lisa Brenan; Aleksandr Andreev; Ofir Cohen; Sasha Pantel; Atanas Kamburov; Davide Cacchiarelli; Nicole S. Persky; Cong Zhu; Mukta Bagul; Eva M. Goetz; Alex B. Burgin; Levi A. Garraway; Gad Getz; Tarjei S. Mikkelsen; Federica Piccioni; David E. Root; Cory M. Johannessen

Tumor-specific genomic information has the potential to guide therapeutic strategies and revolutionize patient treatment. Currently, this approach is limited by an abundance of disease-associated mutants whose biological functions and impacts on therapeutic response are uncharacterized. To begin to address this limitation, we functionally characterized nearly all (99.84%) missense mutants of MAPK1/ERK2, an essential effector of oncogenic RAS and RAF. Using this approach, we discovered rare gain- and loss-of-function ERK2 mutants found in human tumors, revealing that, in the context of this assay, mutational frequency alone cannot identify all functionally impactful mutants. Gain-of-function ERK2 mutants induced variable responses to RAF-, MEK-, and ERK-directed therapies, providing a reference for future treatment decisions. Tumor-associated mutations spatially clustered in two ERK2 effector-recruitment domains yet produced mutants with opposite phenotypes. This approach articulates an allele-characterization framework that can be scaled to meet the goals of genome-guided oncology.


Journal of Clinical Investigation | 2018

CRISPR-Cas9 screen reveals a MYCN -amplified neuroblastoma dependency on EZH2

Liying Chen; Gabriela Alexe; Neekesh V. Dharia; Linda Ross; Amanda Balboni Iniguez; Amy Saur Conway; Emily Jue Wang; Veronica Veschi; Norris Lam; Jun Qi; W. Clay Gustafson; Nicole Nasholm; Francisca Vazquez; Barbara A. Weir; Glenn S. Cowley; Levi D. Ali; Sasha Pantel; Guozhi Jiang; William F. Harrington; Yenarae Lee; Amy Goodale; Rakela Lubonja; John M. Krill-Burger; Robin M. Meyers; Aviad Tsherniak; David E. Root; James E. Bradner; Todd R. Golub; Charles W. M. Roberts; William C. Hahn

Pharmacologically difficult targets, such as MYC transcription factors, represent a major challenge in cancer therapy. For the childhood cancer neuroblastoma, amplification of the oncogene MYCN is associated with high-risk disease and poor prognosis. Here, we deployed genome-scale CRISPR-Cas9 screening of MYCN-amplified neuroblastoma and found a preferential dependency on genes encoding the polycomb repressive complex 2 (PRC2) components EZH2, EED, and SUZ12. Genetic and pharmacological suppression of EZH2 inhibited neuroblastoma growth in vitro and in vivo. Moreover, compared with neuroblastomas without MYCN amplification, MYCN-amplified neuroblastomas expressed higher levels of EZH2. ChIP analysis showed that MYCN binds at the EZH2 promoter, thereby directly driving expression. Transcriptomic and epigenetic analysis, as well as genetic rescue experiments, revealed that EZH2 represses neuronal differentiation in neuroblastoma in a PRC2-dependent manner. Moreover, MYCN-amplified and high-risk primary tumors from patients with neuroblastoma exhibited strong repression of EZH2-regulated genes. Additionally, overexpression of IGFBP3, a direct EZH2 target, suppressed neuroblastoma growth in vitro and in vivo. We further observed strong synergy between histone deacetylase inhibitors and EZH2 inhibitors. Together, these observations demonstrate that MYCN upregulates EZH2, leading to inactivation of a tumor suppressor program in neuroblastoma, and support testing EZH2 inhibitors in patients with MYCN-amplified neuroblastoma.


Clinical Cancer Research | 2017

Abstract B39: Genomic copy number alterations introduce a gene-independent viability bias in CRISPR-Cas9 knock-out screens of cancer cell lines

Robin M. Meyers; Andrew J. Aguirre; Barbara A. Weir; Francisca Vazquez; Cheng-Zhong Zhang; Uri Ben-David; April Cook; Gavin Ha; William F. Harrington; Mihir Doshi; Stanley Gill; Han Xu; Levi D. Ali; Guozhi Jiang; Sasha Pantel; Yenarae Lee; Amy Goodale; Andrew D. Cherniack; Coyin Oh; Gregory V. Kryukov; Glenn S. Cowley; Levi A. Garraway; Kimberly Stegmaier; Charles W. M. Roberts; Todd R. Golub; Matthew Meyerson; David E. Root; Aviad Tsherniak; William C. Hahn

Recent studies have demonstrated the power of CRISPR-Cas9 screening methods for identifying genetic vulnerabilities in cancer cells. As part of a larger effort to generate a comprehensive catalog of vulnerabilities, we performed CRISPR-Cas9 genome-scale loss-of-function screens in 33 cancer cell lines to identify genes essential for proliferation and survival. We found a strong correlation between gene copy number and cell viability after Cas9-targeting. Copy number alterations are extremely prevalent in human cancers and frequently lead to overexpression of driver oncogenes and potential vulnerabilities. Therefore, we sought to identify such genes by investigating the relationship of genomic copy number with essentiality from our screening data. As expected, known oncogenes scored as essential in cell lines harboring amplifications of these genes. However, the scores of all other genes in these amplified regions were also strongly enriched for apparent essentiality, even among unexpressed genes. Furthermore, the infection of cells with sgRNAs targeting Cas9 to non-coding intergenic sequences within regions of high copy number gain also induced this negative effect on cell viability. We observed this effect across multiple different chromosomal structural alterations, including tandem duplications, breakage-fusion-bridge structures, and arm-level gains. More broadly, we found a striking global correlation between cell viability in response to Cas9-targeting and the genomic copy number of the targeted site, even among low-level copy number gain and loss. For example, Cas9-targeting of genes with two copies resulted in, on average, decreased viability relative to Cas9-targeting of genes with only one copy. By examining sgRNAs that target multiple genomic sites, but not within any amplified loci, we found that this cell response to Cas9-targeting correlated strongly with the total number of target sites. Together, these observations indicate that genome targeting by CRISPR-Cas9 elicits a gene-independent anti-proliferative cell response with a severity proportional to the total number of discrete genomic loci targeted. This effect has important practical implications for interpretation of CRISPR-Cas9 screening data and confounds the use of this technology for identification of essential genes in amplified regions. This result illustrates the sensitivity of cancer cells to site-specific DNA damage, which may provide a path to novel therapeutic strategies. Targeting non-essential genes or non-coding intergenic sequences within regions of copy number amplification may reveal cancer-specific vulnerabilities. Citation Format: Robin M. Meyers, Andrew J. Aguirre, Barbara A. Weir, Francisca Vazquez, Cheng-Zhong Zhang, Uri Ben-David, April Cook, Gavin Ha, William F. Harrington, Mihir Doshi, Stanley Gill, Han Xu, Levi D. Ali, Guozhi Jiang, Sasha Pantel, Yenarae Lee, Amy Goodale, Andrew D. Cherniack, Coyin Oh, Gregory Kryukov, Glenn S. Cowley, Levi A. Garraway, Kimberly Stegmaier, Charles W. Roberts, Todd R. Golub, Matthew Meyerson, David E. Root, Aviad Tsherniak, William C. Hahn. Genomic copy number alterations introduce a gene-independent viability bias in CRISPR-Cas9 knock-out screens of cancer cell lines. [abstract]. In: Proceedings of the AACR Precision Medicine Series: Targeting the Vulnerabilities of Cancer; May 16-19, 2016; Miami, FL. Philadelphia (PA): AACR; Clin Cancer Res 2017;23(1_Suppl):Abstract nr B39.


Abstracts: AACR Precision Medicine Series: Targeting the Vulnerabilities of Cancer; May 16-19, 2016; Miami, FL | 2017

Abstract PR02: Towards a Cancer Dependency Map

Aviad Tsherniak; Francisca Vazquez; Barbara A. Weir; Philip Montgomery; Glenn S. Cowley; Stanley Gill; Gregory V. Kryukov; Sasha Pantel; Will Harrington; Mike Burger; Robin M. Meyers; Levi D. Ali; Amy Goodale; Yenarae Lee; Levi A. Garraway; Jesse S. Boehm; David E. Root; Todd R. Golub; William C. Hahn

The mapping of cancer genomes is rapidly approaching completion. The genomic information encoded by individual patients9 tumors should, in principle, provide a guide for predicting acquired cancer dependencies. Unfortunately, while the success of precision cancer genomics hinges on the decoding of such dependencies, we lack the ability to predict dependencies for most individual tumors. The challenge stems from the absence of clinical data relating genotypes with dependencies since most cancer mutations are rare and our arsenal of cancer drugs is incomplete. A comprehensive Cancer Dependency Map comprised of a catalog of genetic and small molecule vulnerabilities across a diverse set of cancers, along with robust statistical models able to predict these vulnerabilities from molecular and genomic features, would provide a roadmap of targets ripe for therapeutic development and would help reveal the mechanisms underlying the emergence of these vulnerabilities. Here, we report progress in creating a Cancer Dependency Map consisting of the following components: 1) Systematic genetic perturbation (RNAi/CRISPR) of over 600 cancer cell models representing a wide range of human cancers and cell lineages using massively parallel genome scale loss-of-function screens. 2) Computational segregation of on- from off-target effects of RNAi enabling the discovery of outlier dependencies. 3) Predictive modeling to discover biomarkers for each dependency. Our results demonstrate that our analytical approach (DEMETER) that models both gene and miRNA-based seed sequence effects effectively segregates on- from off-target effects of shRNAs. We discover 768 preferential dependencies whose suppression decreases viability at a level greater than six standard deviations in at least one of 503 cancer models and 105 such dependencies each present in at least 15 models. We find that 95% of the cancer models screened are strongly sensitive to the suppression of at least one of these dependencies, and that many models have common dependencies so that all models harbor at least one six-sigma dependency out of a set of only 76. Using a custom random forest based predictive modeling framework (ATLANTIS), we discover predictive biomarkers for hundreds of dependencies. These include known and novel vulnerabilities specified by somatic oncogenic alterations, overexpression of genes that specify lineage and differentiation, copy-number driven essentiality, and loss of functionally redundant paralogs. These observations provide a rigorous computational and experimental foundation for the creation of a comprehensive Cancer Dependency Map. Subsampling and projection analyses suggest that over 10,000 genomically characterized cancer cell models will be needed to achieve this important goal. This abstract is also being presented as Poster B43. Citation Format: Aviad Tsherniak, Francisca Vazquez, Barbara Weir, Philip Montgomery, Glenn Cowley, Stanley Gill, Gregory Kryukov, Sasha Pantel, Will Harrington, Mike Burger, Robin Meyers, Levi Ali, Amy Goodale, Yenarae Lee, Levi Garraway, Jesse Boehm, David Root, Todd Golub, William Hahn. Towards a Cancer Dependency Map. [abstract]. In: Proceedings of the AACR Precision Medicine Series: Targeting the Vulnerabilities of Cancer; May 16-19, 2016; Miami, FL. Philadelphia (PA): AACR; Clin Cancer Res 2017;23(1_Suppl):Abstract nr PR02.


Cancer Research | 2016

Abstract 4368: High-throughput phenotyping of lung cancer somatic mutations

Alice H. Berger; Angela N. Brooks; Xiaoyun Wu; Yashaswi Shrestha; Candace R. Chouinard; Federica Piccioni; Mukta Bagul; Atanas Kamburov; Marcin Imielinski; Larson J. Hogstrom; Cong Zhu; Xiaoping Yang; Sasha Pantel; Ryo Sakai; Nathan Kaplan; David E. Root; Rajiv Narayan; Ted Natoli; David L. Lahr; Itay Tirosh; Pablo Tamayo; Gad Getz; Bang Wong; John G. Doench; Aravind Subramanian; Todd R. Golub; Matthew Meyerson; Jesse S. Boehm

Recent cancer genome sequencing and analysis has identified millions of somatic mutations in cancer. However, the functional impact of most variants is poorly understood, limiting the use of this genetic knowledge for clinical decision-making. Here we describe a new high-throughput approach, expression-based variant impact phenotyping (eVIP), which uses gene expression changes to infer somatic mutation impact. We generated a lentiviral expression library representing 53 genes and 194 somatic mutations identified in primary lung adenocarcinomas. Next, we introduced this library into A549 lung adenocarcinoma cells and 96 hours later performed gene expression profiling using Luminex-based L1000 profiling. We built a computational pipeline, eVIP, to compare mutant and wild-type expression signatures to infer whether variants were gain-of-function, change-of-function, loss-of-function, or neutral. Overall, eVIP identified 69% of mutations as impactful whereas 31% appeared functionally neutral. A very high rate, 92%, of missense mutations in the KEAP1 and STK11 tumor suppressor genes were found to inactivate or diminish protein function. As a complementary approach, we assessed which mutations are epistatic to EGFR or capable of initiating xenograft tumor formation in vivo. A subset of the impactful mutations identified by eVIP could induce xenograft tumor formation in mice and/or confer resistance to cellular EGFR inhibition. Among these mutations were 20 rare or non-canonical somatic variants in clinically-actionable or -relevant oncogenes including EGFR S645C, ARAF S214C and S214F, ERBB2 S418T, and PIK3CA E600K. eVIP can, in principle, characterize any genetic variant, independent of prior knowledge of gene function. Further application of eVIP should significantly advance the pace of functional characterization of mutations identified from genome sequencing. Citation Format: Alice H. Berger, Angela N. Brooks, Xiaoyun Wu, Yashaswi Shrestha, Candace Chouinard, Federica Piccioni, Mukta Bagul, Atanas Kamburov, Marcin Imielinski, Larson Hogstrom, Cong Zhu, Xiaoping Yang, Sasha Pantel, Ryo Sakai, Nathan Kaplan, David Root, Rajiv Narayan, Ted Natoli, David Lahr, Itay Tirosh, Pablo Tamayo, Gad Getz, Bang Wong, John Doench, Aravind Subramanian, Todd R. Golub, Matthew Meyerson, Jesse S. Boehm. High-throughput phenotyping of lung cancer somatic mutations. [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 4368.


Cell | 2017

Defining a Cancer Dependency Map

Aviad Tsherniak; Francisca Vazquez; Phil Montgomery; Barbara A. Weir; Gregory V. Kryukov; Glenn S. Cowley; Stanley Gill; William F. Harrington; Sasha Pantel; John M. Krill-Burger; Robin M. Meyers; Levi D. Ali; Amy Goodale; Yenarae Lee; Guozhi Jiang; Jessica Hsiao; William F.J. Gerath; Sara Howell; Erin Merkel; Mahmoud Ghandi; Levi A. Garraway; David E. Root; Todd R. Golub; Jesse S. Boehm; William C. Hahn


Cancer Research | 2018

Abstract 1815: Massively parallel identification of conserved drug resistant mutations in kinases

Nicole S. Persky; Desiree Hernandez; Jonathon Cordova; Amanda Walker; Lisa Brenan; Federica Piccioni; Sasha Pantel; Yenarae Lee; Amy Goodale; Xiaoping Yang; Yoichiro Mitsuishi; Mariana Do Carmo; Cong Zhu; Aleksandr Andreev; David E. Root; Cory M. Johannessen

Collaboration


Dive into the Sasha Pantel'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
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge