Network


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

Hotspot


Dive into the research topics where Vivek K. Arora is active.

Publication


Featured researches published by Vivek K. Arora.


Cancer Cell | 2010

Integrative Genomic Profiling of Human Prostate Cancer

Barry S. Taylor; Nikolaus Schultz; Haley Hieronymus; Anuradha Gopalan; Yonghong Xiao; Brett S. Carver; Vivek K. Arora; Poorvi Kaushik; Ethan Cerami; Boris Reva; Yevgeniy Antipin; Nicholas Mitsiades; Thomas Landers; Igor Dolgalev; John Major; Manda Wilson; Nicholas D. Socci; Alex E. Lash; Adriana Heguy; James A. Eastham; Howard I. Scher; Victor E. Reuter; Peter T. Scardino; Chris Sander; Charles L. Sawyers; William L. Gerald

Annotation of prostate cancer genomes provides a foundation for discoveries that can impact disease understanding and treatment. Concordant assessment of DNA copy number, mRNA expression, and focused exon resequencing in 218 prostate cancer tumors identified the nuclear receptor coactivator NCOA2 as an oncogene in approximately 11% of tumors. Additionally, the androgen-driven TMPRSS2-ERG fusion was associated with a previously unrecognized, prostate-specific deletion at chromosome 3p14 that implicates FOXP1, RYBP, and SHQ1 as potential cooperative tumor suppressors. DNA copy-number data from primary tumors revealed that copy-number alterations robustly define clusters of low- and high-risk disease beyond that achieved by Gleason score. The genomic and clinical outcome data from these patients are now made available as a public resource.


Science | 2009

Development of a Second-Generation Antiandrogen for Treatment of Advanced Prostate Cancer

Chris Tran; Samedy Ouk; Nicola J. Clegg; Yu Chen; Philip A. Watson; Vivek K. Arora; John Wongvipat; Peter Smith-Jones; Dongwon Yoo; Andrew Kwon; Teresa Wasielewska; Derek S. Welsbie; Charlie D. Chen; Celestia S. Higano; Tomasz M. Beer; David T. Hung; Howard I. Scher; Michael E. Jung; Charles L. Sawyers

A Second Act for Antiandrogens Men with advanced prostate cancer are often treated with antiandrogens; drugs that inhibit the activity of male hormones, such as testosterone, that help drive tumor growth. Many of these drugs act by functionally disrupting the androgen receptor (AR), a transcriptional regulator of cell proliferation, but tumors eventually become resistant to the drugs by expressing higher levels of the AR. Tran et al. (p. 787, published online 9 April) have developed a “second-generation” antiandrogen, a thiohydantoin called MDV3100, which binds the AR with high affinity. MDV3100 retains its anticancer activity in cell culture and in mouse models even when AR levels are elevated. The drug appears to act both by inhibiting translocation of the AR into the nucleus and by reducing its transcriptional activity. MDV3100 is being tested in patients with advanced prostate cancer, the first group of which have shown a decline in blood levels of a marker of cancer growth, prostate-specific antigen. A drug that binds to the androgen receptor acts by disrupting its activity in the cell nucleus. Metastatic prostate cancer is treated with drugs that antagonize androgen action, but most patients progress to a more aggressive form of the disease called castration-resistant prostate cancer, driven by elevated expression of the androgen receptor. Here we characterize the diarylthiohydantoins RD162 and MDV3100, two compounds optimized from a screen for nonsteroidal antiandrogens that retain activity in the setting of increased androgen receptor expression. Both compounds bind to the androgen receptor with greater relative affinity than the clinically used antiandrogen bicalutamide, reduce the efficiency of its nuclear translocation, and impair both DNA binding to androgen response elements and recruitment of coactivators. RD162 and MDV3100 are orally available and induce tumor regression in mouse models of castration-resistant human prostate cancer. Of the first 30 patients treated with MDV3100 in a Phase I/II clinical trial, 13 of 30 (43%) showed sustained declines (by >50%) in serum concentrations of prostate-specific antigen, a biomarker of prostate cancer. These compounds thus appear to be promising candidates for treatment of advanced prostate cancer.


Cancer Cell | 2011

Reciprocal Feedback Regulation of PI3K and Androgen Receptor Signaling in PTEN-Deficient Prostate Cancer

Brett S. Carver; Caren Chapinski; John Wongvipat; Haley Hieronymus; Yu Chen; Sarat Chandarlapaty; Vivek K. Arora; Carl Le; Jason A. Koutcher; Howard I. Scher; Peter T. Scardino; Neal Rosen; Charles L. Sawyers

Prostate cancer is characterized by its dependence on androgen receptor (AR) and frequent activation of PI3K signaling. We find that AR transcriptional output is decreased in human and murine tumors with PTEN deletion and that PI3K pathway inhibition activates AR signaling by relieving feedback inhibition of HER kinases. Similarly, AR inhibition activates AKT signaling by reducing levels of the AKT phosphatase PHLPP. Thus, these two oncogenic pathways cross-regulate each other by reciprocal feedback. Inhibition of one activates the other, thereby maintaining tumor cell survival. However, combined pharmacologic inhibition of PI3K and AR signaling caused near-complete prostate cancer regressions in a Pten-deficient murine prostate cancer model and in human prostate cancer xenografts, indicating that both pathways coordinately support survival.


Cell | 2013

Glucocorticoid Receptor Confers Resistance to Antiandrogens by Bypassing Androgen Receptor Blockade

Vivek K. Arora; Emily Schenkein; Rajmohan Murali; Sumit K. Subudhi; John Wongvipat; Minna D. Balbas; Neel Shah; Ling Cai; Chris Logothetis; Deyou Zheng; Charles L. Sawyers

The treatment of advanced prostate cancer has been transformed by novel antiandrogen therapies such as enzalutamide. Here, we identify induction of glucocorticoid receptor (GR) expression as a common feature of drug-resistant tumors in a credentialed preclinical model, a finding also confirmed in patient samples. GR substituted for the androgen receptor (AR) to activate a similar but distinguishable set of target genes and was necessary for maintenance of the resistant phenotype. The GR agonist dexamethasone was sufficient to confer enzalutamide resistance, whereas a GR antagonist restored sensitivity. Acute AR inhibition resulted in GR upregulation in a subset of prostate cancer cells due to relief of AR-mediated feedback repression of GR expression. These findings establish a mechanism of escape from AR blockade through expansion of cells primed to drive AR target genes via an alternative nuclear receptor upon drug exposure.


Cancer Research | 2012

ARN-509: A Novel Antiandrogen for Prostate Cancer Treatment

Nicola J. Clegg; John Wongvipat; James Joseph; Chris Tran; Samedy Ouk; Anna Dilhas; Yu Chen; Kate Grillot; Eric D. Bischoff; Ling Cai; Anna Aparicio; Steven Dorow; Vivek K. Arora; Gang Shao; Jing Qian; Hong Zhao; Guangbin Yang; Chunyan Cao; John Sensintaffar; Teresa Wasielewska; Mark R. Herbert; Celine Bonnefous; Beatrice Darimont; Howard I. Scher; Peter Smith-Jones; Mark Klang; Nicholas D. Smith; Elisa de Stanchina; Nian Wu; Ouathek Ouerfelli

Continued reliance on the androgen receptor (AR) is now understood as a core mechanism in castration-resistant prostate cancer (CRPC), the most advanced form of this disease. While established and novel AR pathway-targeting agents display clinical efficacy in metastatic CRPC, dose-limiting side effects remain problematic for all current agents. In this study, we report the discovery and development of ARN-509, a competitive AR inhibitor that is fully antagonistic to AR overexpression, a common and important feature of CRPC. ARN-509 was optimized for inhibition of AR transcriptional activity and prostate cancer cell proliferation, pharmacokinetics, and in vivo efficacy. In contrast to bicalutamide, ARN-509 lacked significant agonist activity in preclinical models of CRPC. Moreover, ARN-509 lacked inducing activity for AR nuclear localization or DNA binding. In a clinically valid murine xenograft model of human CRPC, ARN-509 showed greater efficacy than MDV3100. Maximal therapeutic response in this model was achieved at 30 mg/kg/d of ARN-509, whereas the same response required 100 mg/kg/d of MDV3100 and higher steady-state plasma concentrations. Thus, ARN-509 exhibits characteristics predicting a higher therapeutic index with a greater potential to reach maximally efficacious doses in man than current AR antagonists. Our findings offer preclinical proof of principle for ARN-509 as a promising therapeutic in both castration-sensitive and castration-resistant forms of prostate cancer.


eLife | 2013

Overcoming mutation-based resistance to antiandrogens with rational drug design

Minna D. Balbas; Michael J. Evans; David J. Hosfield; John Wongvipat; Vivek K. Arora; Philip A. Watson; Yu Chen; Geoffrey L. Greene; Yang Shen; Charles L. Sawyers

The second-generation antiandrogen enzalutamide was recently approved for patients with castration-resistant prostate cancer. Despite its success, the duration of response is often limited. For previous antiandrogens, one mechanism of resistance is mutation of the androgen receptor (AR). To prospectively identify AR mutations that might confer resistance to enzalutamide, we performed a reporter-based mutagenesis screen and identified a novel mutation, F876L, which converted enzalutamide into an AR agonist. Ectopic expression of AR F876L rescued the growth inhibition of enzalutamide treatment. Molecular dynamics simulations performed on antiandrogen–AR complexes suggested a mechanism by which the F876L substitution alleviates antagonism through repositioning of the coactivator recruiting helix 12. This model then provided the rationale for a focused chemical screen which, based on existing antiandrogen scaffolds, identified three novel compounds that effectively antagonized AR F876L (and AR WT) to suppress the growth of prostate cancer cells resistant to enzalutamide. DOI: http://dx.doi.org/10.7554/eLife.00499.001


Nature Reviews Cancer | 2015

Emerging Mechanisms of Resistance to Androgen Receptor Inhibitors in Prostate Cancer

Philip A. Watson; Vivek K. Arora; Charles L. Sawyers

During the past 10 years, preclinical studies implicating sustained androgen receptor (AR) signalling as the primary driver of castration-resistant prostate cancer (CRPC) have led to the development of novel agents targeting the AR pathway that are now in widespread clinical use. These drugs prolong the survival of patients with late-stage prostate cancer but are not curative. In this Review, we highlight emerging mechanisms of acquired resistance to these contemporary therapies, which fall into the three broad categories of restored AR signalling, AR bypass signalling and complete AR independence. This diverse range of resistance mechanisms presents new challenges for long-term disease control, which may be addressable through early use of combination therapies guided by recent insights from genomic landscape studies of CRPC.


Cancer Discovery | 2013

Androgen Receptor Signaling Regulates DNA Repair in Prostate Cancers

William R. Polkinghorn; Joel S. Parker; Man X. Lee; Elizabeth M. Kass; Daniel E. Spratt; Phillip J. Iaquinta; Vivek K. Arora; Wei Feng Yen; Ling Cai; Deyou Zheng; Brett S. Carver; Yu Chen; Philip A. Watson; Neel Shah; Sho Fujisawa; Alexander G. Goglia; Anuradha Gopalan; Haley Hieronymus; John Wongvipat; Peter T. Scardino; Michael J. Zelefsky; Maria Jasin; Jayanta Chaudhuri; Simon N. Powell; Charles L. Sawyers

UNLABELLED We demonstrate that the androgen receptor (AR) regulates a transcriptional program of DNA repair genes that promotes prostate cancer radioresistance, providing a potential mechanism by which androgen deprivation therapy synergizes with ionizing radiation. Using a model of castration-resistant prostate cancer, we show that second-generation antiandrogen therapy results in downregulation of DNA repair genes. Next, we demonstrate that primary prostate cancers display a significant spectrum of AR transcriptional output, which correlates with expression of a set of DNA repair genes. Using RNA-seq and ChIP-seq, we define which of these DNA repair genes are both induced by androgen and represent direct AR targets. We establish that prostate cancer cells treated with ionizing radiation plus androgen demonstrate enhanced DNA repair and decreased DNA damage and furthermore that antiandrogen treatment causes increased DNA damage and decreased clonogenic survival. Finally, we demonstrate that antiandrogen treatment results in decreased classical nonhomologous end-joining. SIGNIFICANCE We demonstrate that the AR regulates a network of DNA repair genes, providing a potential mechanism by which androgen deprivation synergizes with radiotherapy for prostate cancer.


Clinical Cancer Research | 2014

Aggressive Variants of Castration-Resistant Prostate Cancer

Himisha Beltran; Scott A. Tomlins; Ana Aparicio; Vivek K. Arora; David S. Rickman; Gustavo Ayala; Jiaoti Huang; Lawrence D. True; Martin E. Gleave; Howard R. Soule; Christopher J. Logothetis; Mark A. Rubin

A subset of patients with advanced castration-resistant prostate cancer may eventually evolve into an androgen receptor (AR)–independent phenotype, with a clinical picture associated with the development of rapidly progressive disease involving visceral sites and hormone refractoriness, often in the setting of a low or modestly rising serum prostate-specific antigen level. Biopsies performed in such patients may vary, ranging from poorly differentiated carcinomas to mixed adenocarcinoma-small cell carcinomas to pure small cell carcinomas. These aggressive tumors often demonstrate low or absent AR protein expression and, in some cases, express markers of neuroendocrine differentiation. Because tumor morphology is not always predicted by clinical behavior, the terms “anaplastic prostate cancer” or “neuroendocrine prostate cancer” have been used descriptively to describe these rapidly growing clinical features. Patients meeting clinical criteria of anaplastic prostate cancer have been shown to predict for poor prognosis, and these patients may be considered for platinum-based chemotherapy treatment regimens. Therefore, understanding variants within the spectrum of advanced prostate cancer has important diagnostic and treatment implications. Clin Cancer Res; 20(11); 2846–50. ©2014 AACR.


Microbes and Infection | 2002

Nef: agent of cell subversion

Vivek K. Arora; Brenda L. Fredericksen; J. Victor Garcia

Primate lentiviruses encode a small protein designated Nef that has been shown to be a major determinant of virus pathogenicity. Nef regulates multiple host factors in order to optimize the cellular environment for virus replication. The mechanisms by which this small protein modulates distinct host cell properties provide intriguing insight into the intricate interaction between virus and host.

Collaboration


Dive into the Vivek K. Arora's collaboration.

Top Co-Authors

Avatar

John Wongvipat

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Charles L. Sawyers

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Peter T. Scardino

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Yu Chen

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Anuradha Gopalan

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Benjamin W. Fischer-Valuck

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Brian C. Baumann

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Bruce J. Roth

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Gerald L. Andriole

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Howard I. Scher

Memorial Sloan Kettering Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge