Network


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

Hotspot


Dive into the research topics where Rajal B. Shah is active.

Publication


Featured researches published by Rajal B. Shah.


Nature | 2007

Distinct classes of chromosomal rearrangements create oncogenic ETS gene fusions in prostate cancer.

Scott A. Tomlins; Bharathi Laxman; Saravana M. Dhanasekaran; Beth E. Helgeson; Xuhong Cao; David S. Morris; Anjana Menon; Xiaojun Jing; Qi Cao; Bo Han; Jindan Yu; Lei Wang; James E. Montie; Mark A. Rubin; Kenneth J. Pienta; Diane Roulston; Rajal B. Shah; Sooryanarayana Varambally; Rohit Mehra; Arul M. Chinnaiyan

Recently, we identified recurrent gene fusions involving the 5′ untranslated region of the androgen-regulated gene TMPRSS2 and the ETS (E26 transformation-specific) family genes ERG, ETV1 or ETV4 in most prostate cancers. Whereas TMPRSS2–ERG fusions are predominant, fewer TMPRSS2–ETV1 cases have been identified than expected on the basis of the frequency of high (outlier) expression of ETV1 (refs 3–13). Here we explore the mechanism of ETV1 outlier expression in human prostate tumours and prostate cancer cell lines. We identified previously unknown 5′ fusion partners in prostate tumours with ETV1 outlier expression, including untranslated regions from a prostate-specific androgen-induced gene (SLC45A3) and an endogenous retroviral element (HERV-K_22q11.23), a prostate-specific androgen-repressed gene (C15orf21), and a strongly expressed housekeeping gene (HNRPA2B1). To study aberrant activation of ETV1, we identified two prostate cancer cell lines, LNCaP and MDA-PCa 2B, that had ETV1 outlier expression. Through distinct mechanisms, the entire ETV1 locus (7p21) is rearranged to a 1.5-megabase prostate-specific region at 14q13.3–14q21.1 in both LNCaP cells (cryptic insertion) and MDA-PCa 2B cells (balanced translocation). Because the common factor of these rearrangements is aberrant ETV1 overexpression, we recapitulated this event in vitro and in vivo, demonstrating that ETV1 overexpression in benign prostate cells and in the mouse prostate confers neoplastic phenotypes. Identification of distinct classes of ETS gene rearrangements demonstrates that dormant oncogenes can be activated in prostate cancer by juxtaposition to tissue-specific or ubiquitously active genomic loci. Subversion of active genomic regulatory elements may serve as a more generalized mechanism for carcinoma development. Furthermore, the identification of androgen-repressed and insensitive 5′ fusion partners may have implications for the anti-androgen treatment of advanced prostate cancer.


Cancer Research | 2004

Androgen-Independent Prostate Cancer Is a Heterogeneous Group of Diseases Lessons from a Rapid Autopsy Program

Rajal B. Shah; Rohit Mehra; Arul M. Chinnaiyan; Ronglai Shen; Debashis Ghosh; Ming Zhou; Gary R. MacVicar; Soorynarayana Varambally; Jason Harwood; Tarek A. Bismar; Robert Kim; Mark A. Rubin; Kenneth J. Pienta

Understanding the biology of prostate cancer metastasis has been limited by the lack of tissue for study. We studied the clinical data, distribution of prostate cancer involvement, morphology, immunophenotypes, and gene expression from 30 rapid autopsies of men who died of hormone-refractory prostate cancer. A tissue microarray was constructed and quantitatively evaluated for expression of prostate-specific antigen, androgen receptor, chromogranin, synaptophysin, MIB-1, and α-methylacylCoA-racemase markers. Hierarchical clustering of 16 rapid autopsy tumor samples was performed to evaluate the cDNA expression pattern associated with the morphology. Comparisons were made between patients as well as within the same patient. Metastatic hormone-refractory prostate cancer has a heterogeneous morphology, immunophenotype, and genotype, demonstrating that “metastatic disease” is a group of diseases even within the same patient. An appreciation of this heterogeneity is critical to evaluating diagnostic and prognostic biomarkers as well as to designing therapeutic targets for advanced disease.


Proceedings of the National Academy of Sciences of the United States of America | 2008

Temporal activation of p53 by a specific MDM2 inhibitor is selectively toxic to tumors and leads to complete tumor growth inhibition

Sanjeev Shangary; Dongguang Qin; Donna McEachern; Meilan Liu; Rebecca Miller; Su Qiu; Zaneta Nikolovska-Coleska; Ke Ding; Guoping Wang; Jianyong Chen; Denzil Bernard; Jian Zhang; Yipin Lu; Qingyang Gu; Rajal B. Shah; Kenneth J. Pienta; Xiaolan Ling; Sanmao Kang; Ming Guo; Yi Sun; Dajun Yang; Shaomeng Wang

We have designed MI-219 as a potent, highly selective and orally active small-molecule inhibitor of the MDM2–p53 interaction. MI-219 binds to human MDM2 with a Ki value of 5 nM and is 10,000-fold selective for MDM2 over MDMX. It disrupts the MDM2–p53 interaction and activates the p53 pathway in cells with wild-type p53, which leads to induction of cell cycle arrest in all cells and selective apoptosis in tumor cells. MI-219 stimulates rapid but transient p53 activation in established tumor xenograft tissues, resulting in inhibition of cell proliferation, induction of apoptosis, and complete tumor growth inhibition. MI-219 activates p53 in normal tissues with minimal p53 accumulation and is not toxic to animals. MI-219 warrants clinical investigation as a new agent for cancer treatment.


Cancer Research | 2006

TMPRSS2:ETV4 gene fusions define a third molecular subtype of prostate cancer.

Scott A. Tomlins; Rohit Mehra; Daniel R. Rhodes; Lisa Smith; Diane Roulston; Beth E. Helgeson; Xuhong Cao; John T. Wei; Mark A. Rubin; Rajal B. Shah; Arul M. Chinnaiyan

Although common in hematologic and mesenchymal malignancies, recurrent gene fusions have not been well characterized in epithelial carcinomas. Recently, using a novel bioinformatic approach, we identified recurrent gene fusions between TMPRSS2 and the ETS family members ERG or ETV1 in the majority of prostate cancers. Here, we interrogated the expression of all ETS family members in prostate cancer profiling studies and identified marked overexpression of ETV4 in 2 of 98 cases. In one such case, we confirmed the overexpression of ETV4 using quantitative PCR, and by rapid amplification of cDNA ends, quantitative PCR, and fluorescence in situ hybridization, we show that the TMPRSS2 (21q22) and ETV4 (17q21) loci are fused in this case. This result defines a third molecular subtype of prostate cancer and supports the hypothesis that dysregulation of ETS family members through fusions with TMRPSS2 may be an initiating event in prostate cancer development.


Modern Pathology | 2007

Comprehensive assessment of TMPRSS2 and ETS family gene aberrations in clinically localized prostate cancer

Rohit Mehra; Scott A. Tomlins; Ronglai Shen; Owais Nadeem; Lei Wang; John T. Wei; Kenneth J. Pienta; Debashis Ghosh; Mark A. Rubin; Arul M. Chinnaiyan; Rajal B. Shah

Novel recurrent gene fusions between the androgen-regulated gene TMPRSS2 and the ETS family members ERG, ETV1, or ETV4 have been recently identified as a common molecular event in prostate cancer development. We comprehensively analyzed the frequency and risk of disease progression for the TMPRSS2 and ETS family genes rearrangements in a cohort of 96 American men surgically treated for clinically localized prostate cancer. Using three break apart (TMPRSS2, ERG, ETV4) and one fusion (TMPRSS:ETV1) fluorescence in situ hybridization (FISH) assays, we identified rearrangements in TMPRSS2, ERG, ETV1, and ETV4 in 65, 55, 2, and 2% of cases, respectively. Overall, 54 and 2% of cases demonstrated TMPRSS2:ERG and TMPRSS2:ETV1 fusions, respectively. As intronic loss of genomic DNA between TMPRSS2 and ERG has been identified as a mechanism of TMPRSS2:ERG fusion, our assays allowed us to detect deletion of the 3′ end of TMPRSS2 and the 5′ end of ERG in 41 and 39% of cases rearranged for respective genes. Prostate cancers demonstrating TMPRSS2 gene rearrangement were associated with high pathologic stage (P=0.04). Our results confirm that recurrent chromosomal aberrations in TMPRSS2 and/or ETS family members are found in about 70% of prostate cancers. Importantly, we define a novel approach to study these gene fusions and identified cases where TMPRSS2 was rearranged without rearrangement of ERG, ETV1 or ETV4 and cases with ETS family gene rearrangement without TMPRSS2 rearrangement, suggesting that novel 5′ and 3′ partners may be involved in gene fusions in prostate cancer.


Cancer Cell | 2008

The role of SPINK1 in ETS rearrangement-negative prostate cancers

Scott A. Tomlins; Daniel R. Rhodes; Jianjun Yu; Sooryanarayana Varambally; Rohit Mehra; Sven Perner; Francesca Demichelis; Beth E. Helgeson; Bharathi Laxman; David S. Morris; Qi Cao; Xuhong Cao; Ove Andrén; Katja Fall; Laura A. Johnson; John T. Wei; Rajal B. Shah; Hikmat Al-Ahmadie; James A. Eastham; Samson W. Fine; Kristina Hotakainen; Ulf-Håkan Stenman; Alex Tsodikov; William L. Gerald; Hans Lilja; Victor E. Reuter; Phillip W. Kantoff; Peter T. Scardino; Mark A. Rubin; Anders Bjartell

ETS gene fusions have been characterized in a majority of prostate cancers; however, the key molecular alterations in ETS-negative cancers are unclear. Here we used an outlier meta-analysis (meta-COPA) to identify SPINK1 outlier expression exclusively in a subset of ETS rearrangement-negative cancers ( approximately 10% of total cases). We validated the mutual exclusivity of SPINK1 expression and ETS fusion status, demonstrated that SPINK1 outlier expression can be detected noninvasively in urine, and observed that SPINK1 outlier expression is an independent predictor of biochemical recurrence after resection. We identified the aggressive 22RV1 cell line as a SPINK1 outlier expression model and demonstrate that SPINK1 knockdown in 22RV1 attenuates invasion, suggesting a functional role in ETS rearrangement-negative prostate cancers.


Cancer Research | 2008

Characterization of TMPRSS2-ETS Gene Aberrations in Androgen-Independent Metastatic Prostate Cancer

Rohit Mehra; Scott A. Tomlins; Jianjun Yu; Xuhong Cao; Lei Wang; Anjana Menon; Mark A. Rubin; Kenneth J. Pienta; Rajal B. Shah; Arul M. Chinnaiyan

Recurrent gene fusions between the androgen-regulated gene TMPRSS2 and the ETS transcription factor family members ERG, ETV1, and ETV4 have been identified as a critical event in prostate cancer development. In this study, we characterized the prevalence and diversity of these rearrangements in hormone-refractory metastatic prostate cancer. We used a fluorescence in situ hybridization (FISH) split probe strategy to comprehensively evaluate TMPRSS2-ETS aberrations across 97 nonosseous metastatic sites of prostate cancer from 30 rapid autopsies of men who died of androgen-independent disease. Tissue microarrays were constructed representing multiple metastatic sites from each patient, and split signal FISH probes for TMPRSS2, ERG, ETV1, and ETV4 were used to assess for TMPRSS2-ETS rearrangements. In patients exhibiting these aberrations, multiple sites from an individual case harbored the same gene fusion molecular subtype suggesting clonal expansion of disease. The most common prostate cancer gene fusion, TMPRSS2-ERG, can be generated by the mechanism of interstitial deletion (Edel) about 39% to 60% of the time in clinically localized disease. Interestingly, we observed that all of the androgen-independent metastatic prostate cancer sites harboring TMPRSS2-ERG were associated with Edel. These findings suggest that TMPRSS2-ERG with Edel is an aggressive and, in this study, uniformly lethal molecular subtype of prostate cancer associated with androgen-independent disease.


The American Journal of Surgical Pathology | 2008

Clear Cell Papillary Renal Cell Carcinoma : A Distinct Histopathologic and Molecular Genetic Entity

Stefano Gobbo; John N. Eble; David J. Grignon; Guido Martignoni; Gregory T. MacLennan; Rajal B. Shah; Shaobo Zhang; Matteo Brunelli; Liang Cheng

A group of renal tumors composed mainly of cells with clear cytoplasm arranged in papillary patterns and arising in end-stage kidneys has recently been identified. The aim of our study is to investigate the cytogenetic and immunohistochemical phenotypes of these unusual renal tumors, and of morphologically similar tumors arising in kidneys unaffected by end-stage renal disease. Seven tumors from 5 patients (age range: 53 to 64 y, mean: 60 y; 3 men and 2 women) were identified. Sections were obtained from paraffin blocks, including the tumors and adjacent non-neoplastic renal parenchyma. Interphase fluorescence in situ hybridization was performed with centromeric probes for chromosomes 3, 7, 17, Y, and with a subtelomeric probe for 3p25. Immunohistochemistry was performed with antibodies against cytokeratin 7, carbonic anhydrase IX, α-methylacyl-CoA racemase, CD10, and transcription factor E3. Four of the tumors were from patients who did not have end-stage renal disease. One patient had end-stage renal disease and presented with 3 morphologically identical tumors, composed of clear cells arranged in a mixture of cystic and papillary structures. Follow-up data were available from all patients and none showed recurrence or metastasis (mean follow-up: 24 mo). All 7 tumors (ranging from 4 to 50 mm in diameter) were stage pT1. All tumors lacked the gains of chromosome 7 and losses of chromosome Y that are typical of papillary renal cell carcinoma. Only 1 tumor showed gain of chromosome 17. Deletion of 3p, usually seen in clear cell renal cell carcinoma, was not detected. All tumors showed strongly positive immunohistochemical staining for cytokeratin 7 and carbonic anhydrase IX and negative immunostaining with antibodies against α-methylacyl-CoA racemase, CD10, and transcription factor E3. In conclusion, clear cell papillary renal cell carcinoma can arise in otherwise normal kidneys and in kidneys with end-stage renal disease. This tumor has immunophenotypic and genetic profiles distinct from those of either classic papillary or clear cell renal cell carcinoma, and should be considered a distinct entity in the spectrum of renal cell neoplasia.


Clinical Cancer Research | 2008

Characterization of TMPRSS2-ERG fusion high-grade prostatic intraepithelial neoplasia and potential clinical implications.

Juan Miguel Mosquera; Sven Perner; Elizabeth M. Genega; Martin G. Sanda; Matthias D. Hofer; Kirsten D. Mertz; Pamela L. Paris; Jeff Simko; Tarek A. Bismar; Gustavo Ayala; Rajal B. Shah; Massimo Loda; Mark A. Rubin

Purpose: More than 1,300,000 prostate needle biopsies are done annually in the United States with up to 16% incidence of isolated high-grade prostatic intraepithelial neoplasia (HGPIN). HGPIN has low predictive value for identifying prostate cancer on subsequent needle biopsies in prostate-specific antigen–screened populations. In contemporary series, prostate cancer is detected in ∼20% of repeat biopsies following a diagnosis of HGPIN. Further, discrete histologic subtypes of HGPIN with clinical implication in management have not been characterized. The TMPRSS2-ERG gene fusion that has recently been described in prostate cancer has also been shown to occur in a subset of HGPIN. This may have significant clinical implications given that TMPRSS2-ERG fusion prostate cancer is associated with a more aggressive clinical course. Experimental Design: In this study, we assessed a series of HGPIN lesions and paired prostate cancer for the presence of TMPRSS2-ERG gene fusion. Results: Fusion-positive HGPIN was observed in 16% of the 143 number of lesions, and in all instances, the matching cancer shared the same fusion pattern. Sixty percent of TMPRSS2-ERG fusion prostate cancer had fusion-negative HGPIN. Conclusions: Given the more aggressive nature of TMPRSS2-ERG prostate cancer, the findings of this study raise the possibility that gene fusion-positive HGPIN lesions are harbingers of more aggressive disease. To date, pathologic, molecular, and clinical variables do not help stratify which men with HGPIN are at increased risk for a cancer diagnosis. Our results suggest that the detection of isolated TMPRSS2-ERG fusion HGPIN would improve the positive predictive value of finding TMPRSS2-ERG fusion prostate cancer in subsequent biopsies.


Clinical Cancer Research | 2009

Prevalence of TMPRSS2-ERG Fusion Prostate Cancer among Men Undergoing Prostate Biopsy in the United States

Juan Miguel Mosquera; Rohit Mehra; Meredith M. Regan; Sven Perner; Elizabeth M. Genega; Gerri Bueti; Rajal B. Shah; Sandra M. Gaston; Scott A. Tomlins; John T. Wei; Michael Kearney; Laura A. Johnson; Jeffrey Tang; Arul M. Chinnaiyan; Mark A. Rubin; Martin G. Sanda

Purpose: Fusion of the TMPRSS2 prostate-specific gene with the ERG transcription factor is a putatively oncogenic gene rearrangement that is commonly found in prostate cancer tissue from men undergoing prostatectomy. However, the prevalence of the fusion was less common in samples of transurethral resection of the prostate from a Swedish cohort of patients with incidental prostate cancer followed by watchful waiting, raising the question as to whether the high prevalence in prostatectomy specimens reflects selection bias. We sought to determine the prevalence of TMPRSS2-ERG gene fusion among prostate-specific antigen–screened men undergoing prostate biopsy in the United States. Experimental Design: We studied 140 prostate biopsies from the same number of patients for TMPRSS2-ERG fusion status with a fluorescent in situ hybridization assay. One hundred and thirty-four samples (100 cancer and 34 benign) were assessable. Results:ERG gene rearrangement was detected in 46% of prostate biopsies that were found to have prostate cancer and in 0% of benign prostate biopsies (P < 0.0001). Evaluation of morphologic features showed that cribriform growth, blue-tinged mucin, macronucleoli, and collagenous micronodules were significantly more frequent in TMPRSS2-ERG fusion–positive prostate cancer biopsies than gene fusion–negative prostate cancer biopsies (P ≤ 0.04). No significant association with Gleason score was detected. In addition, non-Caucasian patients were less likely to have positive fusion status (P = 0.02). Conclusions: This is the first prospective North American multicenter study to characterize TMPRSS2-ERG prostate cancer prevalence in a cohort of patients undergoing needle biopsy irrespective of whether or not they subsequently undergo prostatectomy. Our results show that this gene rearrangement is common among North American men who have prostate cancer on biopsy, is absent in benign prostate biopsy, and is associated with specific morphologic features. These findings indicate a need for prospective studies to evaluate the relationship of TMPRSS2-ERG rearrangement with clinical course of screening-detected prostate cancer in North American men, and a need for the development of noninvasive screening tests to detect TMPRSS2-ERG rearrangement.

Collaboration


Dive into the Rajal B. Shah's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

John T. Wei

University of Michigan

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kenneth J. Pienta

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Xuhong Cao

University of Michigan

View shared research outputs
Researchain Logo
Decentralizing Knowledge