Network


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

Hotspot


Dive into the research topics where Sivasish Sindiri is active.

Publication


Featured researches published by Sivasish Sindiri.


Cancer Discovery | 2014

Comprehensive Genomic Analysis of Rhabdomyosarcoma Reveals a Landscape of Alterations Affecting a Common Genetic Axis in Fusion-Positive and Fusion-Negative Tumors

Jack F. Shern; Li Chen; Juliann Chmielecki; Jun S. Wei; Rajesh Patidar; Mara Rosenberg; Lauren Ambrogio; Daniel Auclair; Jianjun Wang; Young K. Song; Catherine Tolman; Laura Hurd; Hongling Liao; Shile Zhang; Dominik Bogen; Andrew S. Brohl; Sivasish Sindiri; Daniel Catchpoole; Thomas C. Badgett; Gad Getz; Jaume Mora; James R. Anderson; Stephen X. Skapek; Frederic G. Barr; Matthew Meyerson; Douglas S. Hawkins; Javed Khan

UNLABELLED Despite gains in survival, outcomes for patients with metastatic or recurrent rhabdomyosarcoma remain dismal. In a collaboration between the National Cancer Institute, Childrens Oncology Group, and Broad Institute, we performed whole-genome, whole-exome, and transcriptome sequencing to characterize the landscape of somatic alterations in 147 tumor/normal pairs. Two genotypes are evident in rhabdomyosarcoma tumors: those characterized by the PAX3 or PAX7 fusion and those that lack these fusions but harbor mutations in key signaling pathways. The overall burden of somatic mutations in rhabdomyosarcoma is relatively low, especially in tumors that harbor a PAX3/7 gene fusion. In addition to previously reported mutations in NRAS, KRAS, HRAS, FGFR4, PIK3CA, and CTNNB1, we found novel recurrent mutations in FBXW7 and BCOR, providing potential new avenues for therapeutic intervention. Furthermore, alteration of the receptor tyrosine kinase/RAS/PIK3CA axis affects 93% of cases, providing a framework for genomics-directed therapies that might improve outcomes for patients with rhabdomyosarcoma. SIGNIFICANCE This is the most comprehensive genomic analysis of rhabdomyosarcoma to date. Despite a relatively low mutation rate, multiple genes were recurrently altered, including NRAS, KRAS, HRAS, FGFR4, PIK3CA, CTNNB1, FBXW7, and BCOR. In addition, a majority of rhabdomyosarcoma tumors alter the receptor tyrosine kinase/RAS/PIK3CA axis, providing an opportunity for genomics-guided intervention.


PLOS Genetics | 2014

The genomic landscape of the Ewing Sarcoma family of tumors reveals recurrent STAG2 mutation.

Andrew S. Brohl; David A. Solomon; Wendy W Chang; Jianjun Wang; Young K. Song; Sivasish Sindiri; Rajesh Patidar; Laura Hurd; Li Chen; Jack F. Shern; Hongling Liao; Xinyu Wen; Julia Gerard; Jung-Sik Kim; José Antonio López Guerrero; Isidro Machado; Daniel H. Wai; Piero Picci; Timothy J. Triche; Andrew E. Horvai; Markku Miettinen; Jun S. Wei; Daniel Catchpool; Antonio Llombart-Bosch; Todd Waldman; Javed Khan

The Ewing sarcoma family of tumors (EFT) is a group of highly malignant small round blue cell tumors occurring in children and young adults. We report here the largest genomic survey to date of 101 EFT (65 tumors and 36 cell lines). Using a combination of whole genome sequencing and targeted sequencing approaches, we discover that EFT has a very low mutational burden (0.15 mutations/Mb) but frequent deleterious mutations in the cohesin complex subunit STAG2 (21.5% tumors, 44.4% cell lines), homozygous deletion of CDKN2A (13.8% and 50%) and mutations of TP53 (6.2% and 71.9%). We additionally note an increased prevalence of the BRCA2 K3326X polymorphism in EFT patient samples (7.3%) compared to population data (OR 7.1, p = 0.006). Using whole transcriptome sequencing, we find that 11% of tumors pathologically diagnosed as EFT lack a typical EWSR1 fusion oncogene and that these tumors do not have a characteristic Ewing sarcoma gene expression signature. We identify samples harboring novel fusion genes including FUS-NCATc2 and CIC-FOXO4 that may represent distinct small round blue cell tumor variants. In an independent EFT tissue microarray cohort, we show that STAG2 loss as detected by immunohistochemistry may be associated with more advanced disease (p = 0.15) and a modest decrease in overall survival (p = 0.10). These results significantly advance our understanding of the genomic and molecular underpinnings of Ewing sarcoma and provide a foundation towards further efforts to improve diagnosis, prognosis, and precision therapeutics testing.


Nature Genetics | 2015

Recurrent inactivating RASA2 mutations in melanoma

Rand Arafeh; Nouar Qutob; Rafi Emmanuel; Alona Keren-Paz; Jason Madore; Abdel G. Elkahloun; James S. Wilmott; Jared J. Gartner; Antonella Di Pizio; Sabina Winograd-Katz; Sivasish Sindiri; Ron Rotkopf; Ken Dutton-Regester; Peter A. Johansson; Antonia L. Pritchard; Nicola Waddell; Victoria Hill; Jimmy C. Lin; Yael Hevroni; Steven A. Rosenberg; Javed Khan; Shifra Ben-Dor; Masha Y. Niv; Igor Ulitsky; Graham J. Mann; Richard A. Scolyer; Nicholas K. Hayward; Yardena Samuels

Analysis of 501 melanoma exomes identified RASA2, encoding a RasGAP, as a tumor-suppressor gene mutated in 5% of melanomas. Recurrent loss-of-function mutations in RASA2 were found to increase RAS activation, melanoma cell growth and migration. RASA2 expression was lost in ≥30% of human melanomas and was associated with reduced patient survival. These findings identify RASA2 inactivation as a melanoma driver and highlight the importance of RasGAPs in cancer.


Journal of Clinical Immunology | 2015

Germline CARD11 Mutation in a Patient with Severe Congenital B Cell Lymphocytosis

Andrew S. Brohl; Jeffrey R. Stinson; Helen C. Su; Thomas Badgett; Chester D. Jennings; Gauthaman Sukumar; Sivasish Sindiri; Wei Wang; Lela Kardava; Susan Moir; Clifton L. Dalgard; Jeffrey A. Moscow; Javed Khan; Andrew L. Snow

PurposeActivating germline mutations in CARD11 have recently been linked to a rare genetic disorder associated with congenital B cell lymphocytosis. We describe a patient with a similar clinical phenotype who had a de novo germline G123D CARD11 mutation.MethodsWhole exome sequencing was performed on DNA from the patient and his biological parents. Laboratory studies examined characteristics of the patient’s B and T lymphocytes. A CARD11 cDNA containing the mutation was transfected into a lymphocyte cell line to gain an understanding of its function. RNA sequencing was performed on samples from the patient and from patients with alternate germline CARD11 mutations and differential gene expression analysis was performed.ResultsThe patient had a decade-long history of severe polyclonal B lymphocytosis in the 20,000–90,000 lymphocytes/mm3 range, which was markedly exacerbated by EBV infection and splenectomy at different times. He had a heterozygous germline CARD11 mutation causing a G123D amino acid substitution, which was demonstrated to induce NF-κB activation in unstimulated lymphocytes. In contrast to previous patients with CARD11 mutations, this patient’s B cells exhibited higher expression of several cell cycle progression genes, as well as enhanced proliferation and improved survival following B cell receptor stimulation.ConclusionsThis is the third reported germline and first de novo CARD11 mutation shown to cause congenital B cell lymphocytosis. The mutation was associated with a dramatically greater lymphocytosis than in previously described cases, disproportionate to the level of constitutive NF-κB activation. However, comparative review of the patient’s clinical history, combined with additional genomic and functional analyses, underscore other important variables that may affect pathophysiology or regulate mutant CARD11 function in B cell proliferation and disease. We now refer to these patients as having BENTA disease (B cell Expansion with NF-κB and T cell Anergy).


Clinical Cancer Research | 2016

MultiDimensional ClinOmics for Precision Therapy of Children and Adolescent Young Adults with Relapsed and Refractory Cancer: A Report from the Center for Cancer Research

Wendy Chang; Andrew S. Brohl; Rajesh Patidar; Sivasish Sindiri; Jack F. Shern; Jun S. Wei; Young K. Song; Marielle E. Yohe; Berkley Gryder; Shile Zhang; Kathleen A. Calzone; Nityashree Shivaprasad; Xinyu Wen; Thomas C. Badgett; Markku Miettinen; Kip R. Hartman; James C. League-Pascual; Toby Trahair; Brigitte C. Widemann; Melinda S. Merchant; Rosandra N. Kaplan; Jimmy Lin; Javed Khan

Purpose: We undertook a multidimensional clinical genomics study of children and adolescent young adults with relapsed and refractory cancers to determine the feasibility of genome-guided precision therapy. Experimental Design: Patients with non-central nervous system solid tumors underwent a combination of whole exome sequencing (WES), whole transcriptome sequencing (WTS), and high-density single-nucleotide polymorphism array analysis of the tumor, with WES of matched germline DNA. Clinically actionable alterations were identified as a reportable germline mutation, a diagnosis change, or a somatic event (including a single nucleotide variant, an indel, an amplification, a deletion, or a fusion gene), which could be targeted with drugs in existing clinical trials or with FDA-approved drugs. Results: Fifty-nine patients in 20 diagnostic categories were enrolled from 2010 to 2014. Ages ranged from 7 months to 25 years old. Seventy-three percent of the patients had prior chemotherapy, and the tumors from these patients with relapsed or refractory cancers had a higher mutational burden than that reported in the literature. Thirty patients (51% of total) had clinically actionable mutations, of which 24 (41%) had a mutation that was currently targetable in a clinical trial setting, 4 patients (7%) had a change in diagnosis, and 7 patients (12%) had a reportable germline mutation. Conclusions: We found a remarkably high number of clinically actionable mutations in 51% of the patients, and 12% with significant germline mutations. We demonstrated the clinical feasibility of next-generation sequencing in a diverse population of relapsed and refractory pediatric solid tumors. Clin Cancer Res; 22(15); 3810–20. ©2016 AACR.


Cancer Discovery | 2017

PAX3–FOXO1 Establishes Myogenic Super Enhancers and Confers BET Bromodomain Vulnerability

Berkley E. Gryder; Marielle E. Yohe; Hsien-Chao Chou; Xiaohu Zhang; Joana Marques; Marco Wachtel; Beat W. Schaefer; Nirmalya Sen; Young K. Song; Alberto Gualtieri; Silvia Pomella; Rossella Rota; Abigail Cleveland; Xinyu Wen; Sivasish Sindiri; Jun S. Wei; Frederic G. Barr; Sudipto Das; Thorkell Andresson; Rajarshi Guha; Madhu Lal-Nag; Marc Ferrer; Jack F. Shern; Keji Zhao; Craig J. Thomas; Javed Khan

Alveolar rhabdomyosarcoma is a life-threatening myogenic cancer of children and adolescent young adults, driven primarily by the chimeric transcription factor PAX3-FOXO1. The mechanisms by which PAX3-FOXO1 dysregulates chromatin are unknown. We find PAX3-FOXO1 reprograms the cis-regulatory landscape by inducing de novo super enhancers. PAX3-FOXO1 uses super enhancers to set up autoregulatory loops in collaboration with the master transcription factors MYOG, MYOD, and MYCN. This myogenic super enhancer circuitry is consistent across cell lines and primary tumors. Cells harboring the fusion gene are selectively sensitive to small-molecule inhibition of protein targets induced by, or bound to, PAX3-FOXO1-occupied super enhancers. Furthermore, PAX3-FOXO1 recruits and requires the BET bromodomain protein BRD4 to function at super enhancers, resulting in a complete dependence on BRD4 and a significant susceptibility to BRD inhibition. These results yield insights into the epigenetic functions of PAX3-FOXO1 and reveal a specific vulnerability that can be exploited for precision therapy.Significance: PAX3-FOXO1 drives pediatric fusion-positive rhabdomyosarcoma, and its chromatin-level functions are critical to understanding its oncogenic activity. We find that PAX3-FOXO1 establishes a myoblastic super enhancer landscape and creates a profound subtype-unique dependence on BET bromodomains, the inhibition of which ablates PAX3-FOXO1 function, providing a mechanistic rationale for exploring BET inhibitors for patients bearing PAX-fusion rhabdomyosarcoma. Cancer Discov; 7(8); 884-99. ©2017 AACR.This article is highlighted in the In This Issue feature, p. 783.


eLife | 2017

Myogenic regulatory transcription factors regulate growth in rhabdomyosarcoma

Inês M. Tenente; Madeline Hayes; Myron S. Ignatius; Karin M. McCarthy; Marielle Yohe; Sivasish Sindiri; Berkley Gryder; Mariana L. Oliveira; Ashwin Ramakrishnan; Qin Tang; Eleanor Chen; G. Petur Nielsen; Javed Khan; David M. Langenau

Rhabdomyosarcoma (RMS) is a pediatric malignacy of muscle with myogenic regulatory transcription factors MYOD and MYF5 being expressed in this disease. Consensus in the field has been that expression of these factors likely reflects the target cell of transformation rather than being required for continued tumor growth. Here, we used a transgenic zebrafish model to show that Myf5 is sufficient to confer tumor-propagating potential to RMS cells and caused tumors to initiate earlier and have higher penetrance. Analysis of human RMS revealed that MYF5 and MYOD are mutually-exclusively expressed and each is required for sustained tumor growth. ChIP-seq and mechanistic studies in human RMS uncovered that MYF5 and MYOD bind common DNA regulatory elements to alter transcription of genes that regulate muscle development and cell cycle progression. Our data support unappreciated and dominant oncogenic roles for MYF5 and MYOD convergence on common transcriptional targets to regulate human RMS growth. DOI: http://dx.doi.org/10.7554/eLife.19214.001


Cell Reports | 2017

The NOTCH1/SNAIL1/MEF2C Pathway Regulates Growth and Self-Renewal in Embryonal Rhabdomyosarcoma

Myron S. Ignatius; Madeline Hayes; Riadh Lobbardi; Eleanor Chen; Karin M. McCarthy; Prethish Sreenivas; Zainab Motala; Adam D. Durbin; Aleksey Molodtsov; Sophia Reeder; Alexander Jin; Sivasish Sindiri; Brian C. Beleyea; Deepak Bhere; Matthew S. Alexander; Khalid Shah; Charles Keller; Corinne M. Linardic; Petur Nielsen; David Malkin; Javed Khan; David M. Langenau

Summary Tumor-propagating cells (TPCs) share self-renewal properties with normal stem cells and drive continued tumor growth. However, mechanisms regulating TPC self-renewal are largely unknown, especially in embryonal rhabdomyosarcoma (ERMS)—a common pediatric cancer of muscle. Here, we used a zebrafish transgenic model of ERMS to identify a role for intracellular NOTCH1 (ICN1) in increasing TPCs by 23-fold. ICN1 expanded TPCs by enabling the de-differentiation of zebrafish ERMS cells into self-renewing myf5+ TPCs, breaking the rigid differentiation hierarchies reported in normal muscle. ICN1 also had conserved roles in regulating human ERMS self-renewal and growth. Mechanistically, ICN1 up-regulated expression of SNAIL1, a transcriptional repressor, to increase TPC number in human ERMS and to block muscle differentiation through suppressing MEF2C, a myogenic differentiation transcription factor. Our data implicate the NOTCH1/SNAI1/MEF2C signaling axis as a major determinant of TPC self-renewal and differentiation in ERMS, raising hope of therapeutically targeting this pathway in the future.


Frontiers in Oncology | 2014

Bioinformatic description of immunotherapy targets for pediatric T-cell leukemia and the impact of normal gene sets used for comparison.

Rimas J. Orentas; Jessica Nordlund; Jianbin He; Sivasish Sindiri; Crystal L. Mackall; Terry J. Fry; Javed Khan

Pediatric lymphoid leukemia has the highest cure rate of all pediatric malignancies, yet due to its prevalence, still accounts for the majority of childhood cancer deaths and requires long-term highly toxic therapy. The ability to target B-cell ALL with immunoglobulin-like binders, whether anti-CD22 antibody or anti-CD19 CAR-Ts, has impacted treatment options for some patients. The development of new ways to target B-cell antigens continues at rapid pace. T-cell ALL accounts for up to 20% of childhood leukemia but has yet to see a set of high-value immunotherapeutic targets identified. To find new targets for T-ALL immunotherapy, we employed a bioinformatic comparison to broad normal tissue arrays, hematopoietic stem cells (HSC), and mature lymphocytes, then filtered the results for transcripts encoding plasma membrane proteins. T-ALL bears a core T-cell signature and transcripts encoding TCR/CD3 components and canonical markers of T-cell development predominate, especially when comparison was made to normal tissue or HSC. However, when comparison to mature lymphocytes was also undertaken, we identified two antigens that may drive, or be associated with leukemogenesis; TALLA-1 and hedgehog interacting protein. In addition, TCR subfamilies, CD1, activation and adhesion markers, membrane-organizing molecules, and receptors linked to metabolism and inflammation were also identified. Of these, only CD52, CD37, and CD98 are currently being targeted clinically. This work provides a set of targets to be considered for future development of immunotherapies for T-ALL.


Cancer | 2018

A mutational comparison of adult and adolescent and young adult (AYA) colon cancer: Comparison of Adult & AYA Colon Cancer

James V. Tricoli; Lisa A. Boardman; Rajesh Patidar; Sivasish Sindiri; Jin S. Jang; William D. Walsh; Paul M. McGregor; Corinne E. Camalier; Michele G. Mehaffey; Wayne L. Furman; Armita Bahrami; P. Mickey Williams; Chih Jian Lih; Barbara A. Conley; Javed Khan

It is possible that the relative lack of progress in treatment outcomes among adolescent and young adult (AYA) patients with cancer is caused by a difference in disease biology compared with the corresponding diseases in younger and older individuals. There is evidence that colon cancer is more aggressive and has a poorer prognosis in AYA patients than in older adult patients.

Collaboration


Dive into the Sivasish Sindiri's collaboration.

Top Co-Authors

Avatar

Javed Khan

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Jun S. Wei

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Jack F. Shern

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Young K. Song

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Xinyu Wen

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Shile Zhang

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Berkley E. Gryder

Georgia Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Hongling Liao

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Berkley Gryder

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge