Network


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

Hotspot


Dive into the research topics where Himisha Beltran is active.

Publication


Featured researches published by Himisha Beltran.


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.


Nature Biotechnology | 2013

Development and validation of a clinical cancer genomic profiling test based on massively parallel DNA sequencing

Garrett Michael Frampton; Alex Fichtenholtz; Geoff Otto; Kai Wang; Sean Downing; Jie He; Michael Schnall-Levin; Jared White; Eric M. Sanford; Peter An; James Sun; Frank Juhn; Kristina Brennan; Kiel Iwanik; Ashley Maillet; Jamie Buell; Emily White; Mandy Zhao; Sohail Balasubramanian; Selmira Terzic; Tina Richards; Vera Banning; Lazaro Garcia; Kristen Mahoney; Zac Zwirko; Amy Donahue; Himisha Beltran; Juan Miguel Mosquera; Mark A. Rubin; Snjezana Dogan

As more clinically relevant cancer genes are identified, comprehensive diagnostic approaches are needed to match patients to therapies, raising the challenge of optimization and analytical validation of assays that interrogate millions of bases of cancer genomes altered by multiple mechanisms. Here we describe a test based on massively parallel DNA sequencing to characterize base substitutions, short insertions and deletions (indels), copy number alterations and selected fusions across 287 cancer-related genes from routine formalin-fixed and paraffin-embedded (FFPE) clinical specimens. We implemented a practical validation strategy with reference samples of pooled cell lines that model key determinants of accuracy, including mutant allele frequency, indel length and amplitude of copy change. Test sensitivity achieved was 95–99% across alteration types, with high specificity (positive predictive value >99%). We confirmed accuracy using 249 FFPE cancer specimens characterized by established assays. Application of the test to 2,221 clinical cases revealed clinically actionable alterations in 76% of tumors, three times the number of actionable alterations detected by current diagnostic tests.


Cancer Discovery | 2011

Molecular Characterization of Neuroendocrine Prostate Cancer and Identification of New Drug Targets

Himisha Beltran; David S. Rickman; Kyung Park; Sung Suk Chae; Andrea Sboner; Theresa Y. MacDonald; Yuwei Wang; Karen Sheikh; Stéphane Terry; Scott T. Tagawa; Rajiv Dhir; Joel B. Nelson; Alexandre de la Taille; Yves Allory; Mark Gerstein; Sven Perner; Kenneth J. Pienta; Arul M. Chinnaiyan; Yuzhuo Wang; Colin Collins; Martin Gleave; Francesca Demichelis; David M. Nanus; Mark A. Rubin

UNLABELLED Neuroendocrine prostate cancer (NEPC) is an aggressive subtype of prostate cancer that most commonly evolves from preexisting prostate adenocarcinoma (PCA). Using Next Generation RNA-sequencing and oligonucleotide arrays, we profiled 7 NEPC, 30 PCA, and 5 benign prostate tissue (BEN), and validated findings on tumors from a large cohort of patients (37 NEPC, 169 PCA, 22 BEN) using IHC and FISH. We discovered significant overexpression and gene amplification of AURKA and MYCN in 40% of NEPC and 5% of PCA, respectively, and evidence that that they cooperate to induce a neuroendocrine phenotype in prostate cells. There was dramatic and enhanced sensitivity of NEPC (and MYCN overexpressing PCA) to Aurora kinase inhibitor therapy both in vitro and in vivo, with complete suppression of neuroendocrine marker expression following treatment. We propose that alterations in Aurora kinase A and N-myc are involved in the development of NEPC, and future clinical trials will help determine from the efficacy of Aurora kinase inhibitor therapy. SIGNIFICANCE We report on the largest in-depth molecular analysis of NEPC and provide new insight into molecular events involved in the progression of prostate cancer.


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.).


European Urology | 2013

Targeted Next-generation Sequencing of Advanced Prostate Cancer Identifies Potential Therapeutic Targets and Disease Heterogeneity

Himisha Beltran; Roman Yelensky; Garrett Michael Frampton; Kyung Park; Sean Downing; Theresa Y. MacDonald; Mirna Jarosz; Doron Lipson; Scott T. Tagawa; David M. Nanus; Philip J. Stephens; Juan Miguel Mosquera; Maureen T. Cronin; Mark A. Rubin

BACKGROUND Most personalized cancer care strategies involving DNA sequencing are highly reliant on acquiring sufficient fresh or frozen tissue. It has been challenging to comprehensively evaluate the genome of advanced prostate cancer (PCa) because of limited access to metastatic tissue. OBJECTIVE To demonstrate the feasibility of a novel next-generation sequencing (NGS)-based platform that can be used with archival formalin-fixed paraffin-embedded (FFPE) biopsy tissue to evaluate the spectrum of DNA alterations seen in advanced PCa. DESIGN, SETTING, AND PARTICIPANTS FFPE samples (including archival prostatectomies and prostate needle biopsies) were obtained from 45 patients representing the spectrum of disease: localized PCa, metastatic hormone-naive PCa, and metastatic castration-resistant PCa (CRPC). We also assessed paired primaries and metastases to understand disease heterogeneity and disease progression. INTERVENTION At least 50 ng of tumor DNA was extracted from FFPE samples and used for hybridization capture and NGS using the Illumina HiSeq 2000 platform. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS A total of 3320 exons of 182 cancer-associated genes and 37 introns of 14 commonly rearranged genes were evaluated for genomic alterations. RESULTS AND LIMITATIONS We obtained an average sequencing depth of >900X. Overall, 44% of CRPCs harbored genomic alterations involving the androgen receptor gene (AR), including AR copy number gain (24% of CRPCs) or AR point mutation (20% of CRPCs). Other recurrent mutations included transmembrane protease, serine 2 gene (TMPRSS2):v-ets erythroblastosis virus E26 oncogene homolog (avian) gene (ERG) fusion (44%); phosphatase and tensin homolog gene (PTEN) loss (44%); tumor protein p53 gene (TP53) mutation (40%); retinoblastoma gene (RB) loss (28%); v-myc myelocytomatosis viral oncogene homolog (avian) gene (MYC) gain (12%); and phosphatidylinositol-4,5-bisphosphate 3-kinase, catalytic subunit α gene (PIK3CA) mutation (4%). There was a high incidence of genomic alterations involving key genes important for DNA repair, including breast cancer 2, early onset gene (BRCA2) loss (12%) and ataxia telangiectasia mutated gene (ATM) mutations (8%); these alterations are potentially targetable with poly(adenosine diphosphate-ribose)polymerase inhibitors. A novel and actionable rearrangement involving the v-raf murine sarcoma viral oncogene homolog B1 gene (BRAF) was also detected. CONCLUSIONS This first-in-principle study demonstrates the feasibility of performing in-depth DNA analyses using FFPE tissue and brings new insight toward understanding the genomic landscape within advanced PCa.


Nature Communications | 2014

The oestrogen receptor alpha-regulated lncRNA NEAT1 is a critical modulator of prostate cancer

Dimple Chakravarty; Andrea Sboner; Sujit S. Nair; Eugenia G. Giannopoulou; Ruohan Li; Sven Hennig; Juan Miguel Mosquera; Jonathan Pauwels; Kyung Park; Myriam Kossai; Theresa Y. MacDonald; Jacqueline Fontugne; Nicholas Erho; Ismael A. Vergara; Mercedeh Ghadessi; Elai Davicioni; Robert B. Jenkins; Nallasivam Palanisamy; Zhengming Chen; Shinichi Nakagawa; Tetsuro Hirose; Neil H. Bander; Himisha Beltran; Archa H. Fox; Olivier Elemento; Mark A. Rubin

The androgen receptor (AR) plays a central role in establishing an oncogenic cascade that drives prostate cancer progression. Some prostate cancers escape androgen dependence and are often associated with an aggressive phenotype. The oestrogen receptor alpha (ERα) is expressed in prostate cancers, independent of AR status. However, the role of ERα remains elusive. Using a combination of chromatin immunoprecipitation (ChIP) and RNA-sequencing data, we identified an ERα-specific non-coding transcriptome signature. Among putatively ERα-regulated intergenic long non-coding RNAs (lncRNAs), we identified nuclear enriched abundant transcript 1 (NEAT1) as the most significantly overexpressed lncRNA in prostate cancer. Analysis of two large clinical cohorts also revealed that NEAT1 expression is associated with prostate cancer progression. Prostate cancer cells expressing high levels of NEAT1 were recalcitrant to androgen or AR antagonists. Finally, we provide evidence that NEAT1 drives oncogenic growth by altering the epigenetic landscape of target gene promoters to favour transcription.


Annals of Oncology | 2015

Management of patients with advanced prostate cancer: recommendations of the St Gallen Advanced Prostate Cancer Consensus Conference (APCCC) 2015

Silke Gillessen; Aurelius Omlin; Gerhardt Attard; J. S. De Bono; Karim Fizazi; Susan Halabi; Peter S. Nelson; Oliver Sartor; Matthew R. Smith; Howard R. Soule; H Akaza; Tomasz M. Beer; Himisha Beltran; Arul M. Chinnaiyan; Gedske Daugaard; Ian D. Davis; M. De Santis; Charles G. Drake; Rosalind Eeles; Stefano Fanti; Martin Gleave; Axel Heidenreich; Maha Hussain; Nicholas D. James; Frédéric Lecouvet; Christopher J. Logothetis; Ken Mastris; Sten Nilsson; William Oh; David Olmos

The first St Gallen Advanced Prostate Cancer Consensus Conference (APCCC) Expert Panel identified and reviewed available evidence for the ten most important areas of controversy in advanced prostate cancer management. Recommendations based on expert opinion are presented. Detailed decisions on treatment will involve clinical consideration of disease extent and location, prior treatments, host factors, patient preferences and logistical and economic constraints.


Cancer Research | 2014

High Fidelity Patient-Derived Xenografts for Accelerating Prostate Cancer Discovery and Drug Development

Dong Lin; Alexander W. Wyatt; Hui Xue; Yuzhuo Wang; Xin Dong; Anne Haegert; Rebecca Wu; Sonal Brahmbhatt; Fan Mo; Jong L; Robert H. Bell; Shawn Anderson; Antonio Hurtado-Coll; Ladan Fazli; Sharma M; Himisha Beltran; Mark A. Rubin; Michael E. Cox; Peter W. Gout; Morris J; Goldenberg L; Stanislav Volik; Martin Gleave; Colin Collins

Standardized and reproducible preclinical models that recapitulate the dynamics of prostate cancer are urgently needed. We established a bank of transplantable patient-derived prostate cancer xenografts that capture the biologic and molecular heterogeneity currently confounding prognostication and therapy development. Xenografts preserved the histopathology, genome architecture, and global gene expression of donor tumors. Moreover, their aggressiveness matched patient observations, and their response to androgen withdrawal correlated with tumor subtype. The panel includes the first xenografts generated from needle biopsy tissue obtained at diagnosis. This advance was exploited to generate independent xenografts from different sites of a primary site, enabling functional dissection of tumor heterogeneity. Prolonged exposure of adenocarcinoma xenografts to androgen withdrawal led to castration-resistant prostate cancer, including the first-in-field model of complete transdifferentiation into lethal neuroendocrine prostate cancer. Further analysis of this model supports the hypothesis that neuroendocrine prostate cancer can evolve directly from adenocarcinoma via an adaptive response and yielded a set of genes potentially involved in neuroendocrine transdifferentiation. We predict that these next-generation models will be transformative for advancing mechanistic understanding of disease progression, response to therapy, and personalized oncology.


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.


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.

Collaboration


Dive into the Himisha Beltran'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