Network


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

Hotspot


Dive into the research topics where Nirali M. Patel is active.

Publication


Featured researches published by Nirali M. Patel.


Cancer Discovery | 2013

Metabolic and Functional Genomic Studies Identify Deoxythymidylate Kinase as a target in LKB1 Mutant Lung Cancer

Yan Liu; Kevin Marks; Glenn S. Cowley; Julian Carretero; Qingsong Liu; Thomas J.F. Niel; Chunxiao Xu; Travis J. Cohoon; Peng Gao; Yong Zhang; Zhao Chen; Abigail Altabef; Jeremy H. Tchaicha; Xiaoxu Wang; Sung Choe; Edward M. Driggers; Jianming Zhang; Sean T. Bailey; Norman E. Sharpless; D. Neil Hayes; Nirali M. Patel; Pasi A. Jänne; Nabeel Bardeesy; Jeffrey A. Engelman; Brendan D. Manning; Reuben J. Shaw; John M. Asara; Ralph Scully; Alec C. Kimmelman; Lauren Averett Byers

The LKB1/STK11 tumor suppressor encodes a serine/threonine kinase, which coordinates cell growth, polarity, motility, and metabolism. In non-small cell lung carcinoma, LKB1 is somatically inactivated in 25% to 30% of cases, often concurrently with activating KRAS mutations. Here, we used an integrative approach to define novel therapeutic targets in KRAS-driven LKB1-mutant lung cancers. High-throughput RNA interference screens in lung cancer cell lines from genetically engineered mouse models driven by activated KRAS with or without coincident Lkb1 deletion led to the identification of Dtymk, encoding deoxythymidylate kinase (DTYMK), which catalyzes dTTP biosynthesis, as synthetically lethal with Lkb1 deficiency in mouse and human lung cancer lines. Global metabolite profiling showed that Lkb1-null cells had a striking decrease in multiple nucleotide metabolites as compared with the Lkb1-wild-type cells. Thus, LKB1-mutant lung cancers have deficits in nucleotide metabolism that confer hypersensitivity to DTYMK inhibition, suggesting that DTYMK is a potential therapeutic target in this aggressive subset of tumors.


Cancer Cytopathology | 2016

FNA smears as a potential source of DNA for targeted next-generation sequencing of lung adenocarcinomas

Amanda L. Treece; Nathan D. Montgomery; Nirali M. Patel; Chris J. Civalier; Leslie G. Dodd; Margaret L. Gulley; Jessica K. Booker; Karen E. Weck

Diff‐Quik–stained fine‐needle aspiration (FNA) smears and touch preparations from biopsies represent alternative specimens for molecular testing when cell block or biopsy material is insufficient. This study describes the use of these samples for targeted next‐generation sequencing (NGS) of primary and metastatic lung adenocarcinoma and reports the DNA quality and success rates of FNA smears versus other specimens from 1 year of clinical use.


PLOS ONE | 2015

Combined Targeted DNA Sequencing in Non-Small Cell Lung Cancer (NSCLC) Using UNCseq and NGScopy, and RNA Sequencing Using UNCqeR for the Detection of Genetic Aberrations in NSCLC

Xiaobei Zhao; Anyou Wang; Vonn Walter; Nirali M. Patel; David A. Eberhard; Michele C. Hayward; Ashley H. Salazar; Heejoon Jo; Matthew G. Soloway; Matthew D. Wilkerson; Joel S. Parker; Xiaoying Yin; Guosheng Zhang; Marni B. Siegel; Gary B. Rosson; H. Shelton Earp; Norman E. Sharpless; Margaret L. Gulley; Karen E. Weck; D. Neil Hayes; Stergios J. Moschos

The recent FDA approval of the MiSeqDx platform provides a unique opportunity to develop targeted next generation sequencing (NGS) panels for human disease, including cancer. We have developed a scalable, targeted panel-based assay termed UNCseq, which involves a NGS panel of over 200 cancer-associated genes and a standardized downstream bioinformatics pipeline for detection of single nucleotide variations (SNV) as well as small insertions and deletions (indel). In addition, we developed a novel algorithm, NGScopy, designed for samples with sparse sequencing coverage to detect large-scale copy number variations (CNV), similar to human SNP Array 6.0 as well as small-scale intragenic CNV. Overall, we applied this assay to 100 snap-frozen lung cancer specimens lacking same-patient germline DNA (07–0120 tissue cohort) and validated our results against Sanger sequencing, SNP Array, and our recently published integrated DNA-seq/RNA-seq assay, UNCqeR, where RNA-seq of same-patient tumor specimens confirmed SNV detected by DNA-seq, if RNA-seq coverage depth was adequate. In addition, we applied the UNCseq assay on an independent lung cancer tumor tissue collection with available same-patient germline DNA (11–1115 tissue cohort) and confirmed mutations using assays performed in a CLIA-certified laboratory. We conclude that UNCseq can identify SNV, indel, and CNV in tumor specimens lacking germline DNA in a cost-efficient fashion.


Clinical Cancer Research | 2016

Germline Analysis from Tumor-Germline Sequencing Dyads to identify clinically actionable secondary findings

Bryce A. Seifert; Julianne M. O'Daniel; Krunal Amin; Daniel S. Marchuk; Nirali M. Patel; Joel S. Parker; Alan P. Hoyle; Lisle E. Mose; Andrew Marron; Michele C. Hayward; Christopher Bizon; Kirk C. Wilhelmsen; James P. Evans; H. Shelton Earp; Norman E. Sharpless; D. Neil Hayes; Jonathan S. Berg

Purpose: To evaluate germline variants in hereditary cancer susceptibility genes among unselected cancer patients undergoing tumor–germline sequencing. Experimental Design: Germline sequence data from 439 individuals undergoing tumor–germline dyad sequencing through the LCCC1108/UNCseq™ (NCT01457196) study were analyzed for genetic variants in 36 hereditary cancer susceptibility genes. These variants were analyzed as an exploratory research study to determine whether pathogenic variants exist within the germline of patients undergoing tumor–germline sequencing. Patients were unselected with respect to indicators of hereditary cancer predisposition. Results: Variants indicative of hereditary cancer predisposition were identified in 19 (4.3%) patients. For about half (10/19), these findings represent new diagnostic information with potentially important implications for the patient and their family. The others were previously identified through clinical genetic evaluation secondary to suspicion of a hereditary cancer predisposition. Genes with pathogenic variants included ATM, BRCA1, BRCA2, CDKN2A, and CHEK2. In contrast, a substantial proportion of patients (178, 40.5%) had Variants of Uncertain Significance (VUS), 24 of which had VUS in genes pertinent to the presenting cancer. Another 143 had VUS in other hereditary cancer genes, and 11 had VUS in both pertinent and nonpertinent genes. Conclusions: Germline analysis in tumor–germline sequencing dyads will occasionally reveal significant germline findings that were clinically occult, which could be beneficial for patients and their families. However, given the low yield for unexpected germline variation and the large proportion of patients with VUS results, analysis and return of germline results should adhere to guidelines for secondary findings rather than diagnostic hereditary cancer testing. Clin Cancer Res; 22(16); 4087–94. ©2016 AACR. See related commentary by Mandelker, p. 3987


The Journal of Molecular Diagnostics | 2016

Multi-Institutional FASTQ File Exchange as a Means of Proficiency Testing for Next-Generation Sequencing Bioinformatics and Variant Interpretation

Kurtis D. Davies; Midhat S. Farooqi; Mike Gruidl; Charles E. Hill; Julie Woolworth-Hirschhorn; Heather Jones; Kenneth L. Jones; Anthony M. Magliocco; Midori Mitui; Philip H. O'Neill; Rebecca O'Rourke; Nirali M. Patel; Dahui Qin; Erica Ramos; Michael R. Rossi; Thomas M. Schneider; Geoffrey H. Smith; Linsheng Zhang; Jason Y. Park; Dara L. Aisner

Next-generation sequencing is becoming increasingly common in clinical laboratories worldwide and is revolutionizing clinical molecular testing. However, the large amounts of raw data produced by next-generation sequencing assays and the need for complex bioinformatics analyses present unique challenges. Proficiency testing in clinical laboratories has traditionally been designed to evaluate assays in their entirety; however, it can be alternatively applied to separate assay components. We developed and implemented a multi-institutional proficiency testing approach to directly assess custom bioinformatics and variant interpretation processes. Six clinical laboratories, all of which use the same commercial library preparation kit for next-generation sequencing analysis of tumor specimens, each submitted raw data (FASTQ files) from four samples. These 24 file sets were then deidentified and redistributed to five of the institutions for analysis and interpretation according to their clinically validated approach. Among the laboratories, there was a high rate of concordance in the calling of single-nucleotide variants, in particular those we considered clinically significant (100% concordance). However, there was significant discordance in the calling of clinically significant insertions/deletions, with only two of seven being called by all participating laboratories. Missed calls were addressed by each laboratory to improve their bioinformatics processes. Thus, through our alternative proficiency testing approach, we identified the bioinformatic detection of insertions/deletions as an area of particular concern for clinical laboratories performing next-generation sequencing testing.


JCI insight | 2016

Subtyping sub-Saharan esophageal squamous cell carcinoma by comprehensive molecular analysis

Wenjin Liu; Jeff M. Snell; William R. Jeck; Katherine A. Hoadley; Matthew D. Wilkerson; Joel S. Parker; Nirali M. Patel; Yohannie Mlombe; Gift Mulima; N. George Liomba; Lindsey L. Wolf; Carol G. Shores; Satish Gopal; Norman E. Sharpless

Esophageal squamous cell carcinoma (ESCC) is endemic in regions of sub-Saharan Africa (SSA), where it is the third most common cancer. Here, we describe whole-exome tumor/normal sequencing and RNA transcriptomic analysis of 59 patients with ESCC in Malawi. We observed similar genetic aberrations as reported in Asian and North American cohorts, including mutations of TP53, CDKN2A, NFE2L2, CHEK2, NOTCH1, FAT1, and FBXW7. Analyses for nonhuman sequences did not reveal evidence for infection with HPV or other occult pathogens. Mutational signature analysis revealed common signatures associated with aging, cytidine deaminase activity (APOBEC), and a third signature of unknown origin, but signatures of inhaled tobacco use, aflatoxin and mismatch repair were notably absent. Based on RNA expression analysis, ESCC could be divided into 3 distinct subtypes, which were distinguished by their expression of cell cycle and neural transcripts. This study demonstrates discrete subtypes of ESCC in SSA, and suggests that the endemic nature of this disease reflects exposure to a carcinogen other than tobacco and oncogenic viruses.


Oncologist | 2017

Enhancing Next‐Generation Sequencing‐Guided Cancer Care Through Cognitive Computing

Nirali M. Patel; Vanessa V. Michelini; Jeff M. Snell; Saianand Balu; Alan P. Hoyle; Joel S. Parker; Michele C. Hayward; David A. Eberhard; Ashley H. Salazar; Patrick McNeillie; Jia Xu; Claudia S. Huettner; Takahiko Koyama; Filippo Utro; Kahn Rhrissorrakrai; Raquel Norel; Erhan Bilal; Ajay K. Royyuru; Laxmi Parida; H. Shelton Earp; Juneko E. Grilley-Olson; D. Neil Hayes; Stephen J. Harvey; Norman E. Sharpless; William Y. Kim

Next‐generation sequencing (NGS) has emerged as an affordable and reproducible means to query tumors for somatic genetic anomalies. To help interpret somatic NGS data, many institutions have created a molecular tumor board to analyze the results of NGS and make recommendations. This article evaluates the utility of cognitive computing systems to analyze data for clinical decision‐making.


Applied Immunohistochemistry & Molecular Morphology | 2016

Identification of Human Papillomavirus Infection in Cancer Tissue by Targeted Next-generation Sequencing.

Nathan D. Montgomery; Joel S. Parker; David A. Eberhard; Nirali M. Patel; Karen E. Weck; Norman E. Sharpless; Zhiyuan Hu; David N. Hayes; Margaret L. Gulley

Human papillomaviruses (HPV) are oncogenic DNA viruses implicated in squamous cell carcinomas of several anatomic sites, as well as endocervical adenocarcinomas. Identification of HPV is an actionable finding in some carcinomas, potentially influencing tumor classification, prognosis, and management. We incorporated capture probes for oncogenic HPV strains 16 and 18 into a broader next-generation sequencing (NGS) panel designed to identify actionable mutations in solid malignancies. A total of 21 head and neck, genitourinary, and gynecologic squamous cell carcinomas and endocervical adenocarcinomas were sequenced as part of the UNCSeq project. Using p16 immunohistochemical results as the gold standard, we set a cutoff for proportion of aligned HPV reads that maximized performance of our NGS assay (92% sensitive, 100% specific for HPV). These results suggest that sequencing of oncogenic pathogens can be incorporated into targeted NGS panels, extending the clinical utility of genomic assays.


Clinical Cancer Research | 2018

Identification of clonal hematopoiesis mutations in solid tumor patients undergoing unpaired next-generation sequencing assays

Catherine C. Coombs; Nancy K. Gillis; Xianming Tan; Jonathan S. Berg; Markus Ball; Maria Balasis; Nathan D. Montgomery; Kelly L Bolton; Joel S. Parker; Tania Mesa; Sean J. Yoder; Michele C. Hayward; Nirali M. Patel; Kristy L. Richards; Christine M. Walko; Todd C. Knepper; John T. Soper; Jared Weiss; Juneko E. Grilley-Olson; William Y. Kim; H. Shelton Earp; Ross L. Levine; Elli Papaemmanuil; Ahmet Zehir; D. Neil Hayes; Eric Padron

Purpose: In this era of precision-based medicine, for optimal patient care, results reported from commercial next-generation sequencing (NGS) assays should adequately reflect the burden of somatic mutations in the tumor being sequenced. Here, we sought to determine the prevalence of clonal hematopoiesis leading to possible misattribution of tumor mutation calls on unpaired Foundation Medicine NGS assays. Experimental Design: This was a retrospective cohort study of individuals undergoing NGS of solid tumors from two large cancer centers. We identified and quantified mutations in genes known to be frequently altered in clonal hematopoiesis (DNMT3A, TET2, ASXL1, TP53, ATM, CHEK2, SF3B1, CBL, JAK2) that were returned to physicians on clinical Foundation Medicine reports. For a subset of patients, we explored the frequency of true clonal hematopoiesis by comparing mutations on Foundation Medicine reports with matched blood sequencing. Results: Mutations in genes that are frequently altered in clonal hematopoiesis were identified in 65% (1,139/1,757) of patients undergoing NGS. When excluding TP53, which is often mutated in solid tumors, these events were still seen in 35% (619/1,757) of patients. Utilizing paired blood specimens, we were able to confirm that 8% (18/226) of mutations reported in these genes were true clonal hematopoiesis events. The majority of DNMT3A mutations (64%, 7/11) and minority of TP53 mutations (4%, 2/50) were clonal hematopoiesis. Conclusions: Clonal hematopoiesis mutations are commonly reported on unpaired NGS testing. It is important to recognize clonal hematopoiesis as a possible cause of misattribution of mutation origin when applying NGS findings to a patients care. See related commentary by Pollyea, p. 5790


Breast Cancer Research and Treatment | 2018

LCCC 1025: a phase II study of everolimus, trastuzumab, and vinorelbine to treat progressive HER2-positive breast cancer brain metastases

Amanda E.D. Van Swearingen; Marni B. Siegel; Allison M. Deal; Maria J. Sambade; Alan P. Hoyle; D. Neil Hayes; Heejoon Jo; Paul Little; Elizabeth Claire Dees; Hyman B. Muss; Trevor Augustus Jolly; Timothy M. Zagar; Nirali M. Patel; C. Ryan Miller; Joel S. Parker; J. Keith Smith; Julie Fisher; Nikita Shah; Lisle Nabell; Rita Nanda; Patrick M. Dillon; Vandana G. Abramson; Lisa A. Carey; Carey K. Anders

PurposeHER2 + breast cancer (BC) is an aggressive subtype with high rates of brain metastases (BCBM). Two-thirds of HER2 + BCBM demonstrate activation of the PI3K/mTOR pathway driving resistance to anti-HER2 therapy. This phase II study evaluated everolimus (E), a brain-permeable mTOR inhibitor, trastuzumab (T), and vinorelbine (V) in patients with HER2 + BCBM.Patients and methodsEligible patients had progressive HER2 + BCBM. The primary endpoint was intracranial response rate (RR); secondary objectives were CNS clinical benefit rate (CBR), extracranial RR, time to progression (TTP), overall survival (OS), and targeted sequencing of tumors from enrolled patients. A two-stage design distinguished intracranial RR of 5% versus 20%.Results32 patients were evaluable for toxicity, 26 for efficacy. Intracranial RR was 4% (1 PR). CNS CBR at 6 mos was 27%; at 3 mos 65%. Median intracranial TTP was 3.9 mos (95% CI 2.2–5). OS was 12.2 mos (95% CI 0.6–20.2). Grade 3–4 toxicities included neutropenia (41%), anemia (16%), and stomatitis (16%). Mutations in TP53 and PIK3CA were common in BCBM. Mutations in the PI3K/mTOR pathway were not associated with response. ERBB2 amplification was higher in BCBM compared to primary BC; ERBB2 amplification in the primary BC trended toward worse OS.ConclusionWhile intracranial RR to ETV was low in HER2 + BCBM patients, one-third achieved CNS CBR; TTP/OS was similar to historical control. No new toxicity signals were observed. Further analysis of the genomic underpinnings of BCBM to identify tractable prognostic and/or predictive biomarkers is warranted. Clinical Trial: (NCT01305941).

Collaboration


Dive into the Nirali M. Patel's collaboration.

Top Co-Authors

Avatar

Joel S. Parker

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Michele C. Hayward

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Karen E. Weck

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

D. Neil Hayes

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Norman E. Sharpless

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

David A. Eberhard

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Juneko E. Grilley-Olson

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

David N. Hayes

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

William Y. Kim

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

H. Shelton Earp

University of North Carolina at Chapel Hill

View shared research outputs
Researchain Logo
Decentralizing Knowledge