Network


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

Hotspot


Dive into the research topics where Wassim Abida is active.

Publication


Featured researches published by Wassim Abida.


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.


The New England Journal of Medicine | 2016

Inherited DNA-Repair Gene Mutations in Men with Metastatic Prostate Cancer

Colin C. Pritchard; Joaquin Mateo; Michael F. Walsh; N. De Sarkar; Wassim Abida; Himisha Beltran; Andrea Garofalo; Roman Gulati; S. Carreira; Rosalind Eeles; Olivier Elemento; Mark A. Rubin; Daniel H. Robinson; Robert J. Lonigro; Maha Hussain; Arul M. Chinnaiyan; Jake Vinson; Julie Filipenko; Levi A. Garraway; Mary-Ellen Taplin; Saud H. Aldubayan; Garam Han; M. Beightol; Colm Morrissey; B. Nghiem; Heather H. Cheng; Bruce Montgomery; Tom Walsh; Silvia Casadei; Michael F. Berger

BACKGROUND Inherited mutations in DNA-repair genes such as BRCA2 are associated with increased risks of lethal prostate cancer. Although the prevalence of germline mutations in DNA-repair genes among men with localized prostate cancer who are unselected for family predisposition is insufficient to warrant routine testing, the frequency of such mutations in patients with metastatic prostate cancer has not been established. METHODS We recruited 692 men with documented metastatic prostate cancer who were unselected for family history of cancer or age at diagnosis. We isolated germline DNA and used multiplex sequencing assays to assess mutations in 20 DNA-repair genes associated with autosomal dominant cancer-predisposition syndromes. RESULTS A total of 84 germline DNA-repair gene mutations that were presumed to be deleterious were identified in 82 men (11.8%); mutations were found in 16 genes, including BRCA2 (37 men [5.3%]), ATM (11 [1.6%]), CHEK2 (10 [1.9% of 534 men with data]), BRCA1 (6 [0.9%]), RAD51D (3 [0.4%]), and PALB2 (3 [0.4%]). Mutation frequencies did not differ according to whether a family history of prostate cancer was present or according to age at diagnosis. Overall, the frequency of germline mutations in DNA-repair genes among men with metastatic prostate cancer significantly exceeded the prevalence of 4.6% among 499 men with localized prostate cancer (P<0.001), including men with high-risk disease, and the prevalence of 2.7% in the Exome Aggregation Consortium, which includes 53,105 persons without a known cancer diagnosis (P<0.001). CONCLUSIONS In our multicenter study, the incidence of germline mutations in genes mediating DNA-repair processes among men with metastatic prostate cancer was 11.8%, which was significantly higher than the incidence among men with localized prostate cancer. The frequencies of germline mutations in DNA-repair genes among men with metastatic disease did not differ significantly according to age at diagnosis or family history of prostate cancer. (Funded by Stand Up To Cancer and others.).


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.


Science | 2017

SOX2 promotes lineage plasticity and antiandrogen resistance in TP53- and RB1-deficient prostate cancer

Ping Mu; Zeda Zhang; Matteo Benelli; Wouter R. Karthaus; Elizabeth Hoover; Chi-Chao Chen; John Wongvipat; Sheng-Yu Ku; Dong Gao; Zhen Cao; Neel Shah; Elizabeth J. Adams; Wassim Abida; Philip A. Watson; Davide Prandi; Chun-Hao Huang; Elisa de Stanchina; Scott W. Lowe; Leigh Ellis; Himisha Beltran; Mark A. Rubin; David W. Goodrich; Francesca Demichelis; Charles L. Sawyers

Evading cancer drugs by identity fraud Prostate cancer growth is fueled by male hormones called androgens. Drugs targeting the androgen receptor (AR) are initially efficacious, but most tumors eventually become resistant (see the Perspective by Kelly and Balk). Mu et al. found that prostate cancer cells escaped the effects of androgen deprivation therapy through a change in lineage identity. Functional loss of the tumor suppressors TP53 and RB1 promoted a shift from AR-dependent luminal epithelial cells to AR-independent basal-like cells. In related work, Ku et al. found that prostate cancer metastasis, lineage switching, and drug resistance were driven by the combined loss of the same tumor suppressors and were accompanied by increased expression of the epigenetic regulator Ezh2. Ezh2 inhibitors reversed the lineage switch and restored sensitivity to androgen deprivation therapy in experimental models. Science, this issue p. 84, p. 78; see also p. 29 Prostate cancer cells escape androgen deprivation therapy by morphing into a cell type that does not require androgens. Some cancers evade targeted therapies through a mechanism known as lineage plasticity, whereby tumor cells acquire phenotypic characteristics of a cell lineage whose survival no longer depends on the drug target. We use in vitro and in vivo human prostate cancer models to show that these tumors can develop resistance to the antiandrogen drug enzalutamide by a phenotypic shift from androgen receptor (AR)–dependent luminal epithelial cells to AR-independent basal-like cells. This lineage plasticity is enabled by the loss of TP53 and RB1 function, is mediated by increased expression of the reprogramming transcription factor SOX2, and can be reversed by restoring TP53 and RB1 function or by inhibiting SOX2 expression. Thus, mutations in tumor suppressor genes can create a state of increased cellular plasticity that, when challenged with antiandrogen therapy, promotes resistance through lineage switching.


Cancer Research | 2008

p53-Dependent and p53-Independent Activation of Autophagy by ARF

Wassim Abida; Wei Gu

The ARF tumor suppressor is a crucial component of the cellular response to hyperproliferative signals, including oncogene activation, and functions by inducing a p53-dependent cell growth arrest and apoptosis program. It has recently been reported that the ARF mRNA can produce a smARF isoform that lacks the NH(2)-terminal region required for p53 activation. Overexpression of this isoform can induce autophagy, a cellular process characterized by the formation of cytoplasmic vesicles and the digestion of cellular content, independently of p53. However, the level of this isoform is extremely low in cells, and it remains unclear whether the predominant form of ARF, the full-length protein, is able to activate autophagy. Here, we show that full-length ARF can induce autophagy in 293T cells where p53 is inactivated by viral proteins, and, notably, expression of the NH(2)-terminal region alone, which is required for nucleolar localization, is sufficient for autophagy activation, independently of p53. Given the reported ability of p53 to induce autophagy, we also investigated the role of p53 in ARF-mediated autophagy induction. We found that full-length ARF expression induces p53 activation and promotes autophagy in a p53-positive cell line, and that ARF-mediated autophagy can be abrogated, at least in part, by RNAi-mediated knockdown of p53 in this cellular context. Thus, our findings modify the current view regarding the mechanism of autophagy induction by ARF and suggest an important role for autophagy in tumor suppression via full-length ARF in both p53-dependent and p53-independent manners, depending on cellular context.


JAMA | 2017

Mutation Detection in Patients With Advanced Cancer by Universal Sequencing of Cancer-Related Genes in Tumor and Normal DNA vs Guideline-Based Germline Testing

Diana Mandelker; Liying Zhang; Yelena Kemel; Zsofia K. Stadler; Vijai Joseph; Ahmet Zehir; Nisha Pradhan; Angela G. Arnold; Michael F. Walsh; Yirong Li; Anoop R. Balakrishnan; Aijazuddin Syed; Meera Prasad; Khedoudja Nafa; Maria I. Carlo; Karen A. Cadoo; Meg Sheehan; Megan Harlan Fleischut; Erin E. Salo-Mullen; Magan Trottier; Steven M. Lipkin; Anne Lincoln; Semanti Mukherjee; Vignesh Ravichandran; Roy Cambria; Jesse Galle; Wassim Abida; Marcia E. Arcila; Ryma Benayed; Ronak Shah

Importance Guidelines for cancer genetic testing based on family history may miss clinically actionable genetic changes with established implications for cancer screening or prevention. Objective To determine the proportion and potential clinical implications of inherited variants detected using simultaneous sequencing of the tumor and normal tissue (“tumor-normal sequencing”) compared with genetic test results based on current guidelines. Design, Setting, and Participants From January 2014 until May 2016 at Memorial Sloan Kettering Cancer Center, 10 336 patients consented to tumor DNA sequencing. Since May 2015, 1040 of these patients with advanced cancer were referred by their oncologists for germline analysis of 76 cancer predisposition genes. Patients with clinically actionable inherited mutations whose genetic test results would not have been predicted by published decision rules were identified. Follow-up for potential clinical implications of mutation detection was through May 2017. Exposure Tumor and germline sequencing compared with the predicted yield of targeted germline sequencing based on clinical guidelines. Main Outcomes and Measures Proportion of clinically actionable germline mutations detected by universal tumor-normal sequencing that would not have been detected by guideline-directed testing. Results Of 1040 patients, the median age was 58 years (interquartile range, 50.5-66 years), 65.3% were male, and 81.3% had stage IV disease at the time of genomic analysis, with prostate, renal, pancreatic, breast, and colon cancer as the most common diagnoses. Of the 1040 patients, 182 (17.5%; 95% CI, 15.3%-19.9%) had clinically actionable mutations conferring cancer susceptibility, including 149 with moderate- to high-penetrance mutations; 101 patients tested (9.7%; 95% CI, 8.1%-11.7%) would not have had these mutations detected using clinical guidelines, including 65 with moderate- to high-penetrance mutations. Frequency of inherited mutations was related to case mix, stage, and founder mutations. Germline findings led to discussion or initiation of change to targeted therapy in 38 patients tested (3.7%) and predictive testing in the families of 13 individuals (1.3%), including 6 for whom genetic evaluation would not have been initiated by guideline-based testing. Conclusions and Relevance In this referral population with selected advanced cancers, universal sequencing of a broad panel of cancer-related genes in paired germline and tumor DNA samples was associated with increased detection of individuals with potentially clinically significant heritable mutations over the predicted yield of targeted germline testing based on current clinical guidelines. Knowledge of these additional mutations can help guide therapeutic and preventive interventions, but whether all of these interventions would improve outcomes for patients with cancer or their family members requires further study. Trial Registration clinicaltrials.gov Identifier: NCT01775072


Nature Genetics | 2018

The long tail of oncogenic drivers in prostate cancer.

Joshua Armenia; Stephanie A. Wankowicz; David R. Liu; Jianjiong Gao; Ritika Kundra; Ed Reznik; Walid K. Chatila; Debyani Chakravarty; G. Celine Han; Ilsa Coleman; Bruce Montgomery; Colin C. Pritchard; Colm Morrissey; Christopher E. Barbieri; Himisha Beltran; Andrea Sboner; Zafeiris Zafeiriou; Susana Miranda; Craig M. Bielski; Alexander Penson; Charlotte Tolonen; Franklin W. Huang; Dan R. Robinson; Yi Mi Wu; Robert J. Lonigro; Levi A. Garraway; Francesca Demichelis; Philip W. Kantoff; Mary-Ellen Taplin; Wassim Abida

Comprehensive genomic characterization of prostate cancer has identified recurrent alterations in genes involved in androgen signaling, DNA repair, and PI3K signaling, among others. However, larger and uniform genomic analysis may identify additional recurrently mutated genes at lower frequencies. Here we aggregate and uniformly analyze exome sequencing data from 1,013 prostate cancers. We identify and validate a new class of E26 transformation-specific (ETS)-fusion-negative tumors defined by mutations in epigenetic regulators, as well as alterations in pathways not previously implicated in prostate cancer, such as the spliceosome pathway. We find that the incidence of significantly mutated genes (SMGs) follows a long-tail distribution, with many genes mutated in less than 3% of cases. We identify a total of 97 SMGs, including 70 not previously implicated in prostate cancer, such as the ubiquitin ligase CUL3 and the transcription factor SPEN. Finally, comparing primary and metastatic prostate cancer identifies a set of genomic markers that may inform risk stratification.Meta-analysis of exome sequencing data identifies new recurrently mutated driver genes for prostate cancer. Comparison of primary and metastatic tumors further identifies genomic markers for advanced prostate cancer that may inform risk stratification.


Cell | 2015

Erratum: 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

Dan 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, Victor E. Reuter, Anuradha Gopalan, Jianjiong Gao, Massimo Loda, Rosina T. Lis, Michaela Bowden, Stephen P. Balk, Glenn Gaviola, Carrie Sougnez, Manaswi Gupta, Evan Y. Yu, Elahe A. Mostaghel, Heather H. Cheng, Hyojeong Mulcahy, Lawrence D. True, Stephen R. Plymate, Heidi Dvinge, Roberta Ferraldeschi, Penny Flohr, Susana Miranda, Zafeiris Zafeiriou, Nina Tunariu, Joaquin Mateo, Raquel Perez-Lopez, Francesca Demichelis, Brian D. Robinson, Andrea Sboner, Marc Schiffman, David M. Nanus, Scott T. Tagawa, Alexandros Sigaras, Kenneth W. Eng, Olivier Elemento, Andrea Sboner, Elisabeth I. Heath, Howard I. Scher, Kenneth J. Pienta, Philip Kantoff, Johann S. de Bono, Mark A. Rubin, Peter S. Nelson, Levi A. Garraway, Charles L. Sawyers,* and Arul M. Chinnaiyan* *Correspondence: [email protected] (C.L.S.), [email protected] (A.M.C.) http://dx.doi.org/10.1016/j.cell.2015.06.053


Hematology-oncology Clinics of North America | 2015

First-Line Treatment and Prognostic Factors of Metastatic Bladder Cancer for Platinum-Eligible Patients

Wassim Abida; Dean F. Bajorin; Jonathan E. Rosenberg

Metastatic urothelial carcinoma is primarily a disease of the elderly, with a median overall survival of approximately 15 months. Cisplatin-based combination chemotherapy is standard first-line treatment for eligible patients, with carboplatin-based regimens used as an alternative for patients considered unfit to receive cisplatin. Prognostic models incorporating clinical risk factors have been validated, and molecular characteristics that predict for treatment response are under investigation. This review summarizes the current status of first-line treatment of metastatic urothelial carcinoma in platinum-eligible patients as well as prognostic and predictive models in this disease.


JCO Precision Oncology | 2017

Prospective Genomic Profiling of Prostate Cancer Across Disease States Reveals Germline and Somatic Alterations That May Affect Clinical Decision Making

Wassim Abida; Joshua Armenia; Anuradha Gopalan; Ryan Brennan; Michael D. Walsh; David Barron; Daniel C. Danila; Dana E. Rathkopf; Michael J. Morris; Susan F. Slovin; Brigit McLaughlin; Kristen Rebecca Curtis; David M. Hyman; Jeremy C. Durack; Stephen B. Solomon; Maria E. Arcila; Ahmet Zehir; Aijazuddin Syed; Jianjiong Gao; Debyani Chakravarty; Hebert Alberto Vargas; Mark E. Robson; Joseph Vijai; Kenneth Offit; Mark T.A. Donoghue; Adam Abeshouse; Ritika Kundra; Zachary J. Heins; Alexander Penson; Christopher C. Harris

PURPOSE A long natural history and a predominant osseous pattern of metastatic spread are impediments to the adoption of precision medicine in patients with prostate cancer. To establish the feasibility of clinical genomic profiling in the disease, we performed targeted deep sequencing of tumor and normal DNA from patients with locoregional, metastatic non-castrate, and metastatic castration-resistant prostate cancer (CRPC). METHODS Patients consented to genomic analysis of their tumor and germline DNA. A hybridization capture-based clinical assay was employed to identify single nucleotide variations, small insertions and deletions, copy number alterations and structural rearrangements in over 300 cancer-related genes in tumors and matched normal blood. RESULTS We successfully sequenced 504 tumors from 451 patients with prostate cancer. Potentially actionable alterations were identified in DNA damage repair (DDR), PI3K, and MAP kinase pathways. 27% of patients harbored a germline or a somatic alteration in a DDR gene that may predict for response to PARP inhibition. Profiling of matched tumors from individual patients revealed that somatic TP53 and BRCA2 alterations arose early in tumors from patients who eventually developed metastatic disease. In contrast, comparative analysis across disease states revealed that APC alterations were enriched in metastatic tumors, while ATM alterations were specifically enriched in CRPC. CONCLUSION Through genomic profiling of prostate tumors representing the disease clinical spectrum, we identified a high frequency of potentially actionable alterations and possible drivers of disease initiation, metastasis and castration-resistance. Our findings support the routine use of tumor and germline DNA profiling for patients with advanced prostate cancer, for the purpose of guiding enrollment in targeted clinical trials and counseling families at increased risk of malignancy.

Collaboration


Dive into the Wassim Abida's collaboration.

Top Co-Authors

Avatar

Charles L. Sawyers

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Howard I. Scher

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Joshua Armenia

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Nikolaus Schultz

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Dana E. Rathkopf

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Michael J. Morris

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

John Wongvipat

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Susan F. Slovin

Memorial Sloan Kettering Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge