Network


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

Hotspot


Dive into the research topics where Harshal Abhyankar is active.

Publication


Featured researches published by Harshal Abhyankar.


Blood | 2014

Mutually exclusive recurrent somatic mutations in MAP2K1 and BRAF support a central role for ERK activation in LCH pathogenesis.

Rikhia Chakraborty; Oliver A. Hampton; Xiaoyun Shen; Stephen J. Simko; Albert Shih; Harshal Abhyankar; Karen Phaik Har Lim; Kyle Covington; Lisa R. Trevino; Ninad Dewal; Donna M. Muzny; HarshaVardhan Doddapaneni; Jianhong Hu; Linghua Wang; Philip J. Lupo; M. John Hicks; Diana L. Bonilla; Karen C. Dwyer; Marie Luise Berres; Poulikos I. Poulikakos; Miriam Merad; Kenneth L. McClain; David A. Wheeler; Carl E. Allen; D. Williams Parsons

Langerhans cell histiocytosis (LCH) is a myeloproliferative disorder characterized by lesions composed of pathological CD207(+) dendritic cells with an inflammatory infiltrate. BRAFV600E remains the only recurrent mutation reported in LCH. In order to evaluate the spectrum of somatic mutations in LCH, whole exome sequencing was performed on matched LCH and normal tissue samples obtained from 41 patients. Lesions from other histiocytic disorders, juvenile xanthogranuloma, Erdheim-Chester disease, and Rosai-Dorfman disease were also evaluated. All of the lesions from histiocytic disorders were characterized by an extremely low overall rate of somatic mutations. Notably, 33% (7/21) of LCH cases with wild-type BRAF and none (0/20) with BRAFV600E harbored somatic mutations in MAP2K1 (6 in-frame deletions and 1 missense mutation) that induced extracellular signal-regulated kinase (ERK) phosphorylation in vitro. Single cases of somatic mutations of the mitogen-activated protein kinase (MAPK) pathway genes ARAF and ERBB3 were also detected. The ability of MAPK pathway inhibitors to suppress MAPK kinase and ERK phosphorylation in cell culture and primary tumor models was dependent on the specific LCH mutation. The findings of this study support a model in which ERK activation is a universal end point in LCH arising from pathological activation of upstream signaling proteins.


Journal of Experimental Medicine | 2014

BRAF-V600E expression in precursor versus differentiated dendritic cells defines clinically distinct LCH risk groups

Marie-Luise Berres; Karen Phaik Har Lim; Tricia L. Peters; Jeremy Price; Hitoshi Takizawa; Hélène Salmon; Juliana Idoyaga; Albert Ruzo; Philip J. Lupo; M. John Hicks; Albert Shih; Stephen J. Simko; Harshal Abhyankar; Rikhia Chakraborty; Marylene Leboeuf; Monique F. Beltrao; Sergio A. Lira; Kenneth Matthew Heym; Björn E. Clausen; Venetia Bigley; Matthew Collin; Markus G. Manz; Kenneth L. McClain; Miriam Merad; Carl E. Allen

The Rockefeller University Press


The Journal of Allergy and Clinical Immunology | 2017

Primary immunodeficiency diseases: Genomic approaches delineate heterogeneous Mendelian disorders

Asbjørg Stray-Pedersen; Hanne Sørmo Sorte; Pubudu Saneth Samarakoon; Tomasz Gambin; Ivan K. Chinn; Zeynep Coban Akdemir; Hans Christian Erichsen; Lisa R. Forbes; Shen Gu; Bo Yuan; Shalini N. Jhangiani; Donna M. Muzny; Olaug K. Rødningen; Ying Sheng; Sarah K. Nicholas; Lenora M. Noroski; Filiz O. Seeborg; Carla M. Davis; Debra L. Canter; Emily M. Mace; Timothy J. Vece; Carl E. Allen; Harshal Abhyankar; Philip M. Boone; Christine R. Beck; Wojciech Wiszniewski; Børre Fevang; Pål Aukrust; Geir E. Tjønnfjord; Tobias Gedde-Dahl

30.00 J. Exp. Med. 2014 Vol. 211 No. 4 669-683 www.jem.org/cgi/doi/10.1084/jem.20130977 669 Langerhans cell histiocytosis (LCH) is characterized by inflammatory lesions that include pathological langerin+ DCs. LCH has pleotropic clinical presentations ranging from single lesions cured by curettage to potentially fatal multisystem disease. The first descriptions of LCH, including Hand-Schüller-Christian disease and Letter-Siwe disease, were based on anatomical location and extent of the lesions (Arceci, 1999). The diagnosis of high-risk LCH, defined by involvement of “risk organs” which include BM, liver, and spleen, conferred mortality rates >20%, where patients with disease limited to non-risk organs (low-risk LCH) had nearly 100% survival, CORRESPONDENCE Carl Allen: [email protected] OR Miriam Merad: [email protected]


The Journal of Pediatrics | 2014

Differentiating skin-limited and multisystem langerhans cell histiocytosis

Stephen J. Simko; Benjamin Garmezy; Harshal Abhyankar; Philip J. Lupo; Rikhia Chakraborty; Karen Phaik Har Lim; Albert Shih; M. John Hicks; Teresa S. Wright; Moise L. Levy; Kenneth L. McClain; Carl E. Allen

Background: Primary immunodeficiency diseases (PIDDs) are clinically and genetically heterogeneous disorders thus far associated with mutations in more than 300 genes. The clinical phenotypes derived from distinct genotypes can overlap. Genetic etiology can be a prognostic indicator of disease severity and can influence treatment decisions. Objective: We sought to investigate the ability of whole‐exome screening methods to detect disease‐causing variants in patients with PIDDs. Methods: Patients with PIDDs from 278 families from 22 countries were investigated by using whole‐exome sequencing. Computational copy number variant (CNV) prediction pipelines and an exome‐tiling chromosomal microarray were also applied to identify intragenic CNVs. Analytic approaches initially focused on 475 known or candidate PIDD genes but were nonexclusive and further tailored based on clinical data, family history, and immunophenotyping. Results: A likely molecular diagnosis was achieved in 110 (40%) unrelated probands. Clinical diagnosis was revised in about half (60/110) and management was directly altered in nearly a quarter (26/110) of families based on molecular findings. Twelve PIDD‐causing CNVs were detected, including 7 smaller than 30 Kb that would not have been detected with conventional diagnostic CNV arrays. Conclusion: This high‐throughput genomic approach enabled detection of disease‐related variants in unexpected genes; permitted detection of low‐grade constitutional, somatic, and revertant mosaicism; and provided evidence of a mutational burden in mixed PIDD immunophenotypes.


Gene Therapy | 2013

The immunogenicity of virus-derived 2A sequences in immunocompetent individuals.

Caroline Arber; Harshal Abhyankar; Helen E. Heslop; Malcolm K. Brenner; Hao Liu; Gianpietro Dotti; Barbara Savoldo

OBJECTIVE To identify features associated with multisystem involvement and therapeutic failure in patients with skin Langerhans cell histiocytosis (LCH). STUDY DESIGN We reviewed medical records of 71 consecutive patients with LCH with skin involvement evaluated at Texas Childrens Hospital and analyzed clinical features, laboratory results, and the presence of circulating cells with the BRAF-V600E mutation with respect to initial staging and clinical outcomes. RESULTS Skin disease in patients older than 18 months of age at diagnosis was associated with the presence of multisystem disease (OR, 9.65; 95% CI, 1.17-79.4). Forty percent of patients referred for presumed skin-limited LCH had underlying multisystem involvement, one-half of these with risk-organ involvement. Patients with skin-limited LCH had a 3-year progression-free survival of 89% after initial therapy, and none developed multisystem disease. Patients with skin/multisystem involvement had a 3-year progression-free survival of 44% with vinblastine/prednisone therapy, and risk-organ involvement did not correlate with failure to achieve nonactive disease. Circulating cells with BRAF-V600E were detected at higher frequency in patients with multisystem involvement (8 of 11 skin/multisystem vs 1 of 13 skin-limited; P = .002). CONCLUSION Skin-limited LCH necessitates infrequent therapeutic intervention and has a lower risk of progression relative to skin plus multisystem LCH. The less-aggressive clinical course and lack of circulating cells with the BRAF-V600E mutation in skin-limited LCH suggest a different mechanism of disease origin compared with multisystem or risk-organ disease.


Oncotarget | 2017

Activating MAPK1 (ERK2) mutation in an aggressive case of disseminated juvenile xanthogranuloma

Rikhia Chakraborty; Oliver A. Hampton; Harshal Abhyankar; Daniel Zinn; Amanda Grimes; Brooks Skull; Olive S. Eckstein; Nadia Mahmood; David A. Wheeler; Dolores Lopez-Terrada; Tricia L. Peters; John Hicks; Tarek Elghetany; Robert A. Krance; Poulikos I. Poulikakos; Miriam Merad; Kenneth L. McClain; Carl E. Allen; Williams D. Parsons

Genetic engineering of T cells for adoptive immunotherapy in cancer patients has shown significant promise. To ensure optimal antitumor activity and safety, the simultaneous expression of multiple genes is frequently required, and short viral-derived 2A sequences are increasingly preferred for this purpose. Concerns exist, however, that these virus-derived sequences may induce unwanted immune responses, and thus diminish persistence of the gene-modified cells after adoptive transfer. Whereas such responses were absent in immunocompromised recipients, potential immunogenicity in immunocompetent individuals remains a concern. We now address whether ex vivo T cell responses can be elicited against the most widely used 2A sequences (2A-Thosea asigna virus (TAV) or 2A-equine rhinitis virus (ERAV), specifically) in immunocompetent individuals. We used a potent ex vivo culture system previously validated to induce T cell responses even against weakly immunogenic antigens. Of the sixteen donors tested, only five released very low levels of interferon-γ in response to 2A-TAV peptide mixtures (single peptide specificity in three donors, adjacent self-antigen peptide specificity in one donor and nonspecific reactivity in one donor). None of them produced cytotoxic activity or responded to 2A-ERAV. These results suggest that exposure to viral-derived 2A sequences is unlikely to produce unwanted T cell responses in immunocompetent individuals and further supports their continued use for studies of human gene therapy.


Blood | 2018

Genetic and mechanistic diversity in pediatric hemophagocytic lymphohistiocytosis

Ivan K. Chinn; Olive S. Eckstein; Erin C. Peckham-Gregory; Baruch R. Goldberg; Lisa R. Forbes; Sarah K. Nicholas; Emily M. Mace; Tiphanie P. Vogel; Harshal Abhyankar; Maria I. Diaz; Helen E. Heslop; Robert A. Krance; Caridad Martinez; Trung C. Nguyen; Dalia A. Bashir; Jordana Goldman; Asbjørg Stray-Pedersen; Luis Alberto Pedroza; M. Cecilia Poli; Juan C. Aldave-Becerra; Sean A. McGhee; Waleed Al-Herz; Aghiad Chamdin; Zeynep H. Coban-Akdemir; Shalini N. Jhangiani; Donna M. Muzny; Tram N. Cao; Diana N. Hong; Richard A. Gibbs; James R. Lupski

Juvenile xanthogranuloma (JXG) is a rare histiocytic disorder that is usually benign and self-limiting. We present a case of atypical, aggressive JXG harboring a novel mitogen-activated protein kinase (MAPK) pathway mutation in the MAPK1 gene, which encodes mitogen-activated protein kinase 1 or extracellular signal-regulated 2 (ERK2). Our analysis revealed that the mutation results in constitutive ERK activation that is resistant to BRAF or MEK inhibitors but susceptible to an ERK inhibitor. These data highlight the importance of identifying specific MAPK pathway alterations as part of the diagnostic workup for patients with histiocytic disorders rather than initiating empiric treatment with MEK inhibitors.


Blood | 2017

A genome-wide association study of LCH identifies a variant in SMAD6 associated with susceptibility

Erin C. Peckham-Gregory; Rikhia Chakraborty; Michael E. Scheurer; John W. Belmont; Harshal Abhyankar; Amel G. Sengal; Brooks Scull; Olive S. Eckstein; Daniel Zinn; Louisa Mayer; Albert Shih; Miriam Merad; D. Williams Parsons; Kenneth L. McClain; Philip J. Lupo; Carl E. Allen

The HLH-2004 criteria are used to diagnose hemophagocytic lymphohistiocytosis (HLH), yet concern exists for their misapplication, resulting in suboptimal treatment of some patients. We sought to define the genomic spectrum and associated outcomes of a diverse cohort of children who met the HLH-2004 criteria. Genetic testing was performed clinically or through research-based whole-exome sequencing. Clinical metrics were analyzed with respect to genomic results. Of 122 subjects enrolled over the course of 17 years, 101 subjects received genetic testing. Biallelic familial HLH (fHLH) gene defects were identified in only 19 (19%) and correlated with presentation at younger than 1 year of age (P < .0001). Digenic fHLH variants were observed but lacked statistical support for disease association. In 28 (58%) of 48 subjects, research whole-exome sequencing analyses successfully identified likely molecular explanations, including underlying primary immunodeficiency diseases, dysregulated immune activation and proliferation disorders, and potentially novel genetic conditions. Two-thirds of patients identified by the HLH-2004 criteria had underlying etiologies for HLH, including genetic defects, autoimmunity, and malignancy. Overall survival was 45%, and increased mortality correlated with HLH triggered by infection or malignancy (P < .05). Differences in survival did not correlate with genetic profile or extent of therapy. HLH should be conceptualized as a phenotype of critical illness characterized by toxic activation of immune cells from different underlying mechanisms. In most patients with HLH, targeted sequencing of fHLH genes remains insufficient for identifying pathogenic mechanisms. Whole-exome sequencing, however, may identify specific therapeutic opportunities and affect hematopoietic stem cell transplantation options for these patients.


Cancer | 2018

CNS Langerhans cell histiocytosis: Common hematopoietic origin for LCH-associated neurodegeneration and mass lesions

Kenneth L. McClain; Jennifer Picarsic; Rikhia Chakraborty; Daniel Zinn; Howard Lin; Harshal Abhyankar; Brooks Scull; Albert Shih; Karen Phaik Har Lim; Olive S. Eckstein; Joseph Lubega; Tricia L. Peters; Walter Olea; Thomas Burke; Nabil Ahmed; M. John Hicks; Brandon Tran; Jeremy Jones; Robert C. Dauser; Michael Jeng; Robert A. Baiocchi; Deborah Schiff; Stanton Goldman; Kenneth Matthew Heym; Harry Wilson; Benjamin Carcamo; Ashish Kumar; Carlos Rodriguez-Galindo; Nicholas S. Whipple; Patrick Campbell

To the editor: Langerhans cell histiocytosis (LCH) is a hematologic disorder that presents with a wide spectrum of symptoms, ranging from focal lesions to potentially lethal multiorgan disease, affecting 4 to 8 per million children per year[1][1] and 1 to 2 per million adults per year.[2][2]


Blood | 2016

Alternative genetic mechanisms of BRAF activation in Langerhans cell histiocytosis.

Rikhia Chakraborty; Burke Tm; Oliver A. Hampton; Daniel Zinn; Karen Phaik Har Lim; Harshal Abhyankar; Brooks Scull; Kumar; Kakkar N; David A. Wheeler; Angshumoy Roy; Poulikos I. Poulikakos; Miriam Merad; Kenneth L. McClain; Donald W. Parsons; Carl E. Allen

Central nervous system Langerhans cell histiocytosis (CNS‐LCH) brain involvement may include mass lesions and/or a neurodegenerative disease (LCH‐ND) of unknown etiology. The goal of this study was to define the mechanisms of pathogenesis that drive CNS‐LCH.

Collaboration


Dive into the Harshal Abhyankar's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Carl E. Allen

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Brooks Scull

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Olive S. Eckstein

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Albert Shih

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Daniel Zinn

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Howard Lin

Baylor College of Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge