Network


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

Hotspot


Dive into the research topics where Kanika Arora is active.

Publication


Featured researches published by Kanika Arora.


Journal of Clinical Oncology | 2016

Genetic Determinants of Cisplatin Resistance in Patients With Advanced Germ Cell Tumors

Aditya Bagrodia; Byron H. Lee; William R. Lee; Eugene K. Cha; John P. Sfakianos; Gopa Iyer; Eugene J. Pietzak; Sizhi Paul Gao; Emily C. Zabor; Irina Ostrovnaya; Samuel D. Kaffenberger; Aijazuddin Syed; Maria E. Arcila; R. S. K. Chaganti; Ritika Kundra; Jana Eng; Joseph Hreiki; Vladimir Vacic; Kanika Arora; Dayna Oschwald; Michael F. Berger; Dean F. Bajorin; Manjit S. Bains; Nikolaus Schultz; Victor E. Reuter; Joel Sheinfeld; George J. Bosl; Hikmat Al-Ahmadie; David B. Solit; Darren R. Feldman

Purpose Owing to its exquisite chemotherapy sensitivity, most patients with metastatic germ cell tumors (GCTs) are cured with cisplatin-based chemotherapy. However, up to 30% of patients with advanced GCT exhibit cisplatin resistance, which requires intensive salvage treatment, and have a 50% risk of cancer-related death. To identify a genetic basis for cisplatin resistance, we performed whole-exome and targeted sequencing of cisplatin-sensitive and cisplatin-resistant GCTs. Methods Men with GCT who received a cisplatin-containing chemotherapy regimen and had available tumor tissue were eligible to participate in this study. Whole-exome sequencing or targeted exon-capture-based sequencing was performed on 180 tumors. Patients were categorized as cisplatin sensitive or cisplatin resistant by using a combination of postchemotherapy parameters, including serum tumor marker levels, radiology, and pathology at surgical resection of residual disease. Results TP53 alterations were present exclusively in cisplatin-resistant tumors and were particularly prevalent among primary mediastinal nonseminomas (72%). TP53 pathway alterations including MDM2 amplifications were more common among patients with adverse clinical features, categorized as poor risk according to the International Germ Cell Cancer Collaborative Group (IGCCCG) model. Despite this association, TP53 and MDM2 alterations predicted adverse prognosis independent of the IGCCCG model. Actionable alterations, including novel RAC1 mutations, were detected in 55% of cisplatin-resistant GCTs. Conclusion In GCT, TP53 and MDM2 alterations were associated with cisplatin resistance and inferior outcomes, independent of the IGCCCG model. The finding of frequent TP53 alterations among mediastinal primary nonseminomas may explain the more frequent chemoresistance observed with this tumor subtype. A substantial portion of cisplatin-resistant GCTs harbor actionable alterations, which might respond to targeted therapies. Genomic profiling of patients with advanced GCT could improve current risk stratification and identify novel therapeutic approaches for patients with cisplatin-resistant disease.


Journal of Experimental Medicine | 2016

Integrative genetic analysis of mouse and human AML identifies cooperating disease alleles

Megan Hatlen; Kanika Arora; Vladimir Vacic; Ewa A. Grabowska; Willey Liao; Bridget Riley-Gillis; Dayna Oschwald; Lan Wang; Jacob E. Joergens; Alan H. Shih; Franck Rapaport; Shengqing Gu; Francesca Voza; Takashi Asai; Benjamin G. Neel; Michael G. Kharas; Mithat Gonen; Ross L. Levine; Stephen D. Nimer

Hatlen et al. provide an integrative analysis of the mutational landscape of mouse and human AML and identify functionally relevant cooperation between AML1-ETO and PTPN11 D61Y. Based on these findings, they generate a novel mouse model of t(8;21)+ AML.


Nature Protocols | 2016

Indel variant analysis of short-read sequencing data with Scalpel

Han Fang; Ewa A. Bergmann; Kanika Arora; Vladimir Vacic; Michael C. Zody; Ivan Iossifov; Jason O'Rawe; Yiyang Wu; Laura Jimenez Barron; Julie Rosenbaum; Michael Ronemus; Yoon-ha Lee; Zihua Wang; Esra Dikoglu; Vaidehi Jobanputra; Gholson J. Lyon; Michael Wigler; Michael C. Schatz; Giuseppe Narzisi

As the second most common type of variation in the human genome, insertions and deletions (indels) have been linked to many diseases, but the discovery of indels of more than a few bases in size from short-read sequencing data remains challenging. Scalpel (http://scalpel.sourceforge.net) is an open-source software for reliable indel detection based on the microassembly technique. It has been successfully used to discover mutations in novel candidate genes for autism, and it is extensively used in other large-scale studies of human diseases. This protocol gives an overview of the algorithm and describes how to use Scalpel to perform highly accurate indel calling from whole-genome and whole-exome sequencing data. We provide detailed instructions for an exemplary family-based de novo study, but we also characterize the other two supported modes of operation: single-sample and somatic analysis. Indel normalization, visualization and annotation of the mutations are also illustrated. Using a standard server, indel discovery and characterization in the exonic regions of the example sequencing data can be completed in ∼5 h after read mapping.


Bioinformatics | 2016

Conpair: concordance and contamination estimator for matched tumor–normal pairs

Ewa A. Bergmann; Bo-Juen Chen; Kanika Arora; Vladimir Vacic; Michael C. Zody

Abstract Motivation: Sequencing of matched tumor and normal samples is the standard study design for reliable detection of somatic alterations. However, even very low levels of cross-sample contamination significantly impact calling of somatic mutations, because contaminant germline variants can be incorrectly interpreted as somatic. There are currently no sequence-only based methods that reliably estimate contamination levels in tumor samples, which frequently display copy number changes. As a solution, we developed Conpair, a tool for detection of sample swaps and cross-individual contamination in whole-genome and whole-exome tumor–normal sequencing experiments. Results: On a ladder of in silico contaminated samples, we demonstrated that Conpair reliably measures contamination levels as low as 0.1%, even in presence of copy number changes. We also estimated contamination levels in glioblastoma WGS and WXS tumor–normal datasets from TCGA and showed that they strongly correlate with tumor–normal concordance, as well as with the number of germline variants called as somatic by several widely-used somatic callers. Availability and Implementation: The method is available at: https://github.com/nygenome/conpair. Contact: [email protected] or [email protected] Supplementary information: Supplementary data are available at Bioinformatics online.


Nature Genetics | 2017

PGBD5 promotes site-specific oncogenic mutations in human tumors

Anton Henssen; Richard Koche; Jiali Zhuang; Eileen Jiang; Casie Reed; Amy Eisenberg; Eric Still; Ian Macarthur; Elias Rodríguez-Fos; Santiago Gonzalez; Montserrat Puiggròs; Andrew N. Blackford; Christopher E. Mason; Elisa de Stanchina; Mithat Gonen; Anne Katrin Emde; Minita Shah; Kanika Arora; Catherine Reeves; Nicholas D. Socci; Elizabeth J. Perlman; Cristina R. Antonescu; Charles W. M. Roberts; Hanno Steen; Elizabeth Mullen; David Torrents; Zhiping Weng; Scott A. Armstrong; Alex Kentsis

Genomic rearrangements are a hallmark of human cancers. Here, we identify the piggyBac transposable element derived 5 (PGBD5) gene as encoding an active DNA transposase expressed in the majority of childhood solid tumors, including lethal rhabdoid tumors. Using assembly-based whole-genome DNA sequencing, we found previously undefined genomic rearrangements in human rhabdoid tumors. These rearrangements involved PGBD5-specific signal (PSS) sequences at their breakpoints and recurrently inactivated tumor-suppressor genes. PGBD5 was physically associated with genomic PSS sequences that were also sufficient to mediate PGBD5-induced DNA rearrangements in rhabdoid tumor cells. Ectopic expression of PGBD5 in primary immortalized human cells was sufficient to promote cell transformation in vivo. This activity required specific catalytic residues in the PGBD5 transposase domain as well as end-joining DNA repair and induced structural rearrangements with PSS breakpoints. These results define PGBD5 as an oncogenic mutator and provide a plausible mechanism for site-specific DNA rearrangements in childhood and adult solid tumors.


Neurology Genetics | 2017

Comparing sequencing assays and human-machine analyses in actionable genomics for glioblastoma.

Kazimierz O. Wrzeszczynski; Mayu O. Frank; Takahiko Koyama; Kahn Rhrissorrakrai; Nicolas Robine; Filippo Utro; Anne-Katrin Emde; Bo-Juen Chen; Kanika Arora; Minita Shah; Vladimir Vacic; Raquel Norel; Erhan Bilal; Ewa A. Bergmann; Julia M. Vogel; Jeffrey N. Bruce; Andrew B. Lassman; Peter Canoll; Christian Grommes; Steve Harvey; Laxmi Parida; Vanessa V. Michelini; Michael C. Zody; Vaidehi Jobanputra; Ajay K. Royyuru; Robert B. Darnell

Objective: To analyze a glioblastoma tumor specimen with 3 different platforms and compare potentially actionable calls from each. Methods: Tumor DNA was analyzed by a commercial targeted panel. In addition, tumor-normal DNA was analyzed by whole-genome sequencing (WGS) and tumor RNA was analyzed by RNA sequencing (RNA-seq). The WGS and RNA-seq data were analyzed by a team of bioinformaticians and cancer oncologists, and separately by IBM Watson Genomic Analytics (WGA), an automated system for prioritizing somatic variants and identifying drugs. Results: More variants were identified by WGS/RNA analysis than by targeted panels. WGA completed a comparable analysis in a fraction of the time required by the human analysts. Conclusions: The development of an effective human-machine interface in the analysis of deep cancer genomic datasets may provide potentially clinically actionable calls for individual patients in a more timely and efficient manner than currently possible. ClinicalTrials.gov identifier: NCT02725684.


Nature Genetics | 2017

Erratum: PGBD5 promotes site-specific oncogenic mutations in human tumors

Anton Henssen; Richard Koche; Jiali Zhuang; Eileen Jiang; Casie Reed; Amy Eisenberg; Eric Still; Ian Macarthur; Elias Rodríguez-Fos; Santiago Gonzalez; Montserrat Puiggròs; Andrew N. Blackford; Christopher E. Mason; Elisa de Stanchina; Mithat Gonen; Anne-Katrin Emde; Minita Shah; Kanika Arora; Catherine Reeves; Nicholas D. Socci; Elizabeth J. Perlman; Cristina R. Antonescu; Charles W. M. Roberts; Hanno Steen; Elizabeth Mullen; David Torrents; Zhiping Weng; Scott A. Armstrong; Alex Kentsis

Nat. Genet.; doi:10.1038/ng.3866; corrected online 24 May 2017 In the version of this article initially published online, the affiliations for Jiali Zhuang listed an incorrect present address instead of an equal contribution. The error has been corrected in the print, PDF and HTML versions of this article.


The Journal of Molecular Diagnostics | 2018

Analytical Validation of Clinical Whole-Genome and Transcriptome Sequencing of Patient-Derived Tumors: Clinical Application of Whole-Genome Sequencing for Reporting Targetable Variants in Cancer

Kazimierz O. Wrzeszczynski; Vanessa Felice; Avinash Abhyankar; Lukasz Kozon; Heather Geiger; Dina Manaa; Ferrah London; Dino Robinson; Xiaolan Fang; David Lin; Michelle Lamendola-Essel; Depinder Khaira; Esra Dikoglu; Anne-Katrin Emde; Nicolas Robine; Minita Shah; Kanika Arora; Olca Basturk; Umesh Bhanot; Alex Kentsis; Mahesh Mansukhani; Govind Bhagat; Vaidehi Jobanputra

We developed and validated a clinical whole-genome and transcriptome sequencing (WGTS) assay that provides a comprehensive genomic profile of a patients tumor. The ability to fully capture the mappable genome with sufficient sequencing coverage to precisely call DNA somatic single nucleotide variants, insertions/deletions, copy number variants, structural variants, and RNA gene fusions was analyzed. New York States Department of Health next-generation DNA sequencing guidelines were expanded for establishing performance validation applicable to whole-genome and transcriptome sequencing. Whole-genome sequencing laboratory protocols were validated for the Illumina HiSeq X Ten platform and RNA sequencing for Illumina HiSeq2500 platform for fresh or frozen and formalin-fixed, paraffin-embedded tumor samples. Various bioinformatics tools were also tested, and CIs for sensitivity and specificity thresholds in calling clinically significant somatic aberrations were determined. The validation was performed on a set of 125 tumor normal pairs. RNA sequencing was performed to call fusions and to confirm the DNA variants or exonic alterations. Here, we present our results and WGTS standards for variant allele frequency, reproducibility, analytical sensitivity, and present limit of detection analysis for single nucleotide variant calling, copy number identification, and structural variants. We show that The New York Genome Center WGTS clinical assay can provide a comprehensive patient variant discovery approach suitable for directed oncologic therapeutic applications.


Cancer Research | 2016

Abstract 4497: NYGC glioblastoma clinical outcomes pilot study: Discovering therapeutic potential in glioblastoma through integrative genomics

Kazimierz O. Wrzeszczynski; Nicolas Robine; Vladimir Vacic; Anne-Katrin Emde; Bo-Juen Chen; Will Liao; Kanika Arora; Minita Shah; Ewa Grabowska; Vanessa Felice; Esra Dikoglu; Catherine Reeves; Mayu O. Frank; Vaidehi Jobanputra; Michael C. Zody; Toby Bloom; Robert B. Darnell

Current adjuvant therapeutic options for the treatment of Glioblastoma (GBM) are often determined by limited histological information. Additionally, most GBM clinical trials for targeted chemotherapeutic agents do not distinguish the genetic mutational tumor profiles of the patients recruited and have failed to reach successful treatment endpoints. The New York Genome Center (NYGC) has undertaken a glioblastoma clinical sequencing outcome pilot study to better determine personal treatment options for patients with GBM using integrated genomic data. During the initial phase of this study for 2015 NYGC has performed whole genome sequencing (WGS) on 10 primary GBM tumor-normal pairs, analyzed each patient9s tumor for single nucleotide variants, structural variants and copy number alterations. In addition RNA sequencing and a DNA methylation assay were also performed on several of the patients. The patient9s genomic profile was then compared to a database of known targeted therapeutic approaches. A final tumor board composed of NYGC scientists, GBM consortium scientists and treating oncologists then reviewed all data prior to identifying a final therapeutic strategy. Data from the first 10 patients revealed RB1 variants to be predominant in half of the patients. SNV9s in NF1 or PIK3R1 were also discovered in 4 out of 10 samples. Remaining lower frequency variants occurred in TP53, PDGFRA, PTEN, PIK3CA, ERBB3, SMO, STAG2, ACVR1, NFKB1 and JAK3. Analysis of copy number alterations resulted in 8 of 10 patients containing the characteristic chromosome 7 amplification combined with chromosome 10 deletion, affecting EGFR and PTEN, respectively. Extreme amplification with potential double minute structural variation of EGFR containing the A289V mutation was observed in 2 of 10 samples. Two samples contained a potentially targetable over-amplification of the PDGFRA/KIT/KDR chromosome 4 locus. Predominant deletions resided in CDKN2A, ESR2, PTEN and FLT3. Hemizygous deletions of RB1 combined with RB1 nonsense or missense variants were observed in 4 samples. To date, RNA sequencing was performed on 5 patient samples. Most strikingly the combination of DNA and RNA sequencing revealed the presence of a putative activating MET exon skipping event in the extracellular domain. This MET variant was considered as a potential targetable variant. Therapeutic options resulting from WGS genomic profiles were the PI3K inhibitor BKM120 (60%), half of these had an additional aberration in MET and were recommended for the combinatorial trial NCT01870726. Drug recommendations for the treatment of GBM based on specific N = 1 patient genomic profiles were also made for nilotinib, vismodegib and palbociclib. Here, we present the first phase of the NYGC GBM clinical outcome study demonstrating how patient WGS information can provide more precise therapeutic options in the treatment of glioblastoma. Citation Format: Kazimierz O. Wrzeszczynski, Nicolas Robine, Vladimir Vacic, Anne-Katrin Emde, Bo-Juen Chen, Will Liao, Kanika Arora, Minita Shah, Ewa A. Grabowska, Vanessa Felice, Esra Dikoglu, Catherine Reeves, Mayu Frank, Vaidehi Jobanputra, Michael C. Zody, Toby Bloom, Robert B. Darnell. NYGC glioblastoma clinical outcomes pilot study: Discovering therapeutic potential in glioblastoma through integrative genomics. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 4497.


bioRxiv | 2018

Genome-wide somatic variant calling using localized colored de Bruijn graphs

Giuseppe Narzisi; André Corvelo; Kanika Arora; Ewa A. Bergmann; Minita Shah; Rajeeva Musunuri; Anne-Katrin Emde; Nicolas Robine; Vladimir Vacic; Michael C. Zody

Collaboration


Dive into the Kanika Arora's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Vaidehi Jobanputra

Columbia University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Alex Kentsis

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Eric Still

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Esra Dikoglu

Cold Spring Harbor Laboratory

View shared research outputs
Researchain Logo
Decentralizing Knowledge