Network


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

Hotspot


Dive into the research topics where Krishna L. Kanchi is active.

Publication


Featured researches published by Krishna L. Kanchi.


Cell | 2012

GENOMIC LANDSCAPE OF NON-SMALL CELL LUNG CANCER IN SMOKERS AND NEVER SMOKERS

Ramaswamy Govindan; Li Ding; Malachi Griffith; Janakiraman Subramanian; Nathan D. Dees; Krishna L. Kanchi; Christopher A. Maher; Robert S. Fulton; Lucinda Fulton; John W. Wallis; Ken Chen; Jason Walker; Sandra A. McDonald; Ron Bose; David M. Ornitz; Dong Hai Xiong; Ming You; David J. Dooling; Mark A. Watson; Elaine R. Mardis; Richard Wilson

We report the results of whole-genome and transcriptome sequencing of tumor and adjacent normal tissue samples from 17 patients with non-small cell lung carcinoma (NSCLC). We identified 3,726 point mutations and more than 90 indels in the coding sequence, with an average mutation frequency more than 10-fold higher in smokers than in never-smokers. Novel alterations in genes involved in chromatin modification and DNA repair pathways were identified, along with DACH1, CFTR, RELN, ABCB5, and HGF. Deep digital sequencing revealed diverse clonality patterns in both never-smokers and smokers. All validated EFGR and KRAS mutations were present in the founder clones, suggesting possible roles in cancer initiation. Analysis revealed 14 fusions, including ROS1 and ALK, as well as novel metabolic enzymes. Cell-cycle and JAK-STAT pathways are significantly altered in lung cancer, along with perturbations in 54 genes that are potentially targetable with currently available drugs.


Nature Communications | 2014

Integrated analysis of germline and somatic variants in ovarian cancer

Krishna L. Kanchi; Kimberly J. Johnson; Charles Lu; Michael D. McLellan; Mark D. M. Leiserson; Michael C. Wendl; Qunyuan Zhang; Daniel C. Koboldt; Mingchao Xie; Cyriac Kandoth; Joshua F. McMichael; Matthew A. Wyczalkowski; David E. Larson; Heather K. Schmidt; Christopher A. Miller; Robert S. Fulton; Paul T. Spellman; Elaine R. Mardis; Todd E. Druley; Timothy A. Graubert; Paul J. Goodfellow; Benjamin J. Raphael; Richard Wilson; Li Ding

We report the first large-scale exome-wide analysis of the combined germline-somatic landscape in ovarian cancer. Here we analyze germline and somatic alterations in 429 ovarian carcinoma cases and 557 controls. We identify 3,635 high confidence, rare truncation and 22,953 missense variants with predicted functional impact. We find germline truncation variants and large deletions across Fanconi pathway genes in 20% of cases. Enrichment of rare truncations is shown in BRCA1, BRCA2, and PALB2. Additionally, we observe germline truncation variants in genes not previously associated with ovarian cancer susceptibility (NF1, MAP3K4, CDKN2B, and MLL3). Evidence for loss of heterozygosity was found in 100% and 76% of cases with germline BRCA1 and BRCA2 truncations respectively. Germline-somatic interaction analysis combined with extensive bioinformatics annotation identifies 237 candidate functional germline truncation and missense variants, including 2 pathogenic BRCA1 and 1 TP53 deleterious variants. Finally, integrated analyses of germline and somatic variants identify significantly altered pathways, including the Fanconi, MAPK, and MLL pathways.


Blood | 2015

Genomic analysis of germ line and somatic variants in familial myelodysplasia/acute myeloid leukemia

Jane E. Churpek; Khateriaa Pyrtel; Krishna L. Kanchi; Jin Shao; Daniel C. Koboldt; Christopher A. Miller; Dong Shen; Robert S. Fulton; Michelle O'Laughlin; Catrina C. Fronick; Iskra Pusic; Geoffrey L. Uy; Evan M. Braunstein; Mark Levis; Julie A. Ross; Kevin Elliott; Sharon Heath; Allan Jiang; Peter Westervelt; John F. DiPersio; Daniel C. Link; Matthew J. Walter; John S. Welch; Richard Wilson; Timothy J. Ley; Lucy A. Godley; Timothy A. Graubert

Familial clustering of myelodysplastic syndromes (MDSs) and acute myeloid leukemia (AML) can be caused by inherited factors. We screened 59 individuals from 17 families with 2 or more biological relatives with MDS/AML for variants in 12 genes with established roles in predisposition to MDS/AML, and identified a pathogenic germ line variant in 5 families (29%). Extending the screen with a panel of 264 genes that are recurrently mutated in de novo AML, we identified rare, nonsynonymous germ line variants in 4 genes, each segregating with MDS/AML in 2 families. Somatic mutations are required for progression to MDS/AML in these familial cases. Using a combination of targeted and exome sequencing of tumor and matched normal samples from 26 familial MDS/AML cases and asymptomatic carriers, we identified recurrent frameshift mutations in the cohesin-associated factor PDS5B, co-occurrence of somatic ASXL1 mutations with germ line GATA2 mutations, and recurrent mutations in other known MDS/AML drivers. Mutations in genes that are recurrently mutated in de novo AML were underrepresented in the familial MDS/AML cases, although the total number of somatic mutations per exome was the same. Lastly, clonal skewing of hematopoiesis was detected in 67% of young, asymptomatic RUNX1 carriers, providing a potential biomarker that could be used for surveillance in these high-risk families.


Nature Communications | 2015

Patterns and functional implications of rare germline variants across 12 cancer types

Charles Lu; Mingchao Xie; Michael C. Wendl; Jiayin Wang; Michael D. McLellan; Mark D. M. Leiserson; Kuan-lin Huang; Matthew A. Wyczalkowski; Reyka Jayasinghe; Tapahsama Banerjee; Jie Ning; Piyush Tripathi; Qunyuan Zhang; Beifang Niu; Kai Ye; Heather K. Schmidt; Robert S. Fulton; Joshua F. McMichael; Prag Batra; Cyriac Kandoth; Maheetha Bharadwaj; Daniel C. Koboldt; Christopher A. Miller; Krishna L. Kanchi; James M. Eldred; David E. Larson; John S. Welch; Ming You; Bradley A. Ozenberger; Ramaswamy Govindan

Large-scale cancer sequencing data enable discovery of rare germline cancer susceptibility variants. Here we systematically analyse 4,034 cases from The Cancer Genome Atlas cancer cases representing 12 cancer types. We find that the frequency of rare germline truncations in 114 cancer-susceptibility-associated genes varies widely, from 4% (acute myeloid leukaemia (AML)) to 19% (ovarian cancer), with a notably high frequency of 11% in stomach cancer. Burden testing identifies 13 cancer genes with significant enrichment of rare truncations, some associated with specific cancers (for example, RAD51C, PALB2 and MSH6 in AML, stomach and endometrial cancers, respectively). Significant, tumour-specific loss of heterozygosity occurs in nine genes (ATM, BAP1, BRCA1/2, BRIP1, FANCM, PALB2 and RAD51C/D). Moreover, our homology-directed repair assay of 68 BRCA1 rare missense variants supports the utility of allelic enrichment analysis for characterizing variants of unknown significance. The scale of this analysis and the somatic-germline integration enable the detection of rare variants that may affect individual susceptibility to tumour development, a critical step toward precision medicine.


PLOS ONE | 2014

Clonal Architectures and Driver Mutations in Metastatic Melanomas

Li Ding; Minjung Kim; Krishna L. Kanchi; Nathan D. Dees; Charles Lu; Malachi Griffith; David Fenstermacher; Hyeran Sung; Christopher A. Miller; Brian D. Goetz; Michael C. Wendl; Obi L. Griffith; Lynn A. Cornelius; Gerald P. Linette; Joshua F. McMichael; Vernon K. Sondak; Ryan C. Fields; Timothy J. Ley; James J. Mulé; Richard Wilson; Jeffrey S. Weber

To reveal the clonal architecture of melanoma and associated driver mutations, whole genome sequencing (WGS) and targeted extension sequencing were used to characterize 124 melanoma cases. Significantly mutated gene analysis using 13 WGS cases and 15 additional paired extension cases identified known melanoma genes such as BRAF, NRAS, and CDKN2A, as well as a novel gene EPHA3, previously implicated in other cancer types. Extension studies using tumors from another 96 patients discovered a large number of truncation mutations in tumor suppressors (TP53 and RB1), protein phosphatases (e.g., PTEN, PTPRB, PTPRD, and PTPRT), as well as chromatin remodeling genes (e.g., ASXL3, MLL2, and ARID2). Deep sequencing of mutations revealed subclones in the majority of metastatic tumors from 13 WGS cases. Validated mutations from 12 out of 13 WGS patients exhibited a predominant UV signature characterized by a high frequency of C->T transitions occurring at the 3′ base of dipyrimidine sequences while one patient (MEL9) with a hypermutator phenotype lacked this signature. Strikingly, a subclonal mutation signature analysis revealed that the founding clone in MEL9 exhibited UV signature but the secondary clone did not, suggesting different mutational mechanisms for two clonal populations from the same tumor. Further analysis of four metastases from different geographic locations in 2 melanoma cases revealed phylogenetic relationships and highlighted the genetic alterations responsible for differential drug resistance among metastatic tumors. Our study suggests that clonal evaluation is crucial for understanding tumor etiology and drug resistance in melanoma.


PLOS Medicine | 2016

Tumor Evolution in Two Patients with Basal-like Breast Cancer: A Retrospective Genomics Study of Multiple Metastases

Katherine A. Hoadley; Marni B. Siegel; Krishna L. Kanchi; Christopher A. Miller; Li Ding; Wei Zhao; Xiaping He; Joel S. Parker; Michael C. Wendl; Robert S. Fulton; Ryan Demeter; Richard Wilson; Lisa A. Carey; Charles M. Perou; Elaine R. Mardis

Background Metastasis is the main cause of cancer patient deaths and remains a poorly characterized process. It is still unclear when in tumor progression the ability to metastasize arises and whether this ability is inherent to the primary tumor or is acquired well after primary tumor formation. Next-generation sequencing and analytical methods to define clonal heterogeneity provide a means for identifying genetic events and the temporal relationships between these events in the primary and metastatic tumors within an individual. Methods and Findings We performed DNA whole genome and mRNA sequencing on two primary tumors, each with either four or five distinct tissue site-specific metastases, from two individuals with triple-negative/basal-like breast cancers. As evidenced by their case histories, each patient had an aggressive disease course with abbreviated survival. In each patient, the overall gene expression signatures, DNA copy number patterns, and somatic mutation patterns were highly similar across each primary tumor and its associated metastases. Almost every mutation found in the primary was found in a metastasis (for the two patients, 52/54 and 75/75). Many of these mutations were found in every tumor (11/54 and 65/75, respectively). In addition, each metastasis had fewer metastatic-specific events and shared at least 50% of its somatic mutation repertoire with the primary tumor, and all samples from each patient grouped together by gene expression clustering analysis. TP53 was the only mutated gene in common between both patients and was present in every tumor in this study. Strikingly, each metastasis resulted from multiclonal seeding instead of from a single cell of origin, and few of the new mutations, present only in the metastases, were expressed in mRNAs. Because of the clinical differences between these two patients and the small sample size of our study, the generalizability of these findings will need to be further examined in larger cohorts of patients. Conclusions Our findings suggest that multiclonal seeding may be common amongst basal-like breast cancers. In these two patients, mutations and DNA copy number changes in the primary tumors appear to have had a biologic impact on metastatic potential, whereas mutations arising in the metastases were much more likely to be passengers.


Oncotarget | 2016

Inactivation of RASA1 promotes melanoma tumorigenesis via R-Ras activation

Hyeran Sung; Krishna L. Kanchi; Xue Wang; Kristen S. Hill; Jane L. Messina; Ji-Hyun Lee; Young Chul Kim; Nathan D. Dees; Li Ding; Jamie K. Teer; Shengyu Yang; Amod A. Sarnaik; Vernon K. Sondak; James J. Mulé; Richard Wilson; Jeffrey S. Weber; Minjung Kim

Inactivation of Ras GTPase activating proteins (RasGAPs) can activate Ras, increasing the risk for tumor development. Utilizing a melanoma whole genome sequencing (WGS) data from 13 patients, we identified two novel, clustered somatic missense mutations (Y472H and L481F) in RASA1 (RAS p21 protein activator 1, also called p120RasGAP). We have shown that wild type RASA1, but not identified mutants, suppresses soft agar colony formation and tumor growth of BRAF mutated melanoma cell lines via its RasGAP activity toward R-Ras (related RAS viral (r-ras) oncogene homolog) isoform. Moreover, R-Ras increased and RASA1 suppressed Ral-A activation among Ras downstream effectors. In addition to mutations, loss of RASA1 expression was frequently observed in metastatic melanoma samples on melanoma tissue microarray (TMA) and a low level of RASA1 mRNA expression was associated with decreased overall survival in melanoma patients with BRAF mutations. Thus, these data support that RASA1 is inactivated by mutation or by suppressed expression in melanoma and that RASA1 plays a tumor suppressive role by inhibiting R-Ras, a previously less appreciated member of the Ras small GTPases.


Genomics data | 2015

Genomic analysis to define molecular basis of aggressiveness in a mouse model of oral cancer

Varun Chalivendra; Krishna L. Kanchi; Michael D. Onken; Ashley E. Winkler; Elaine R. Mardis; Ravindra Uppaluri

To investigate the molecular basis underlying aggressive behavior in oral squamous cell carcinoma (OSCC), our laboratory developed a carcinogen-induced mouse oral cancer (MOC) cell line model that encompasses the growth and metastasis spectrum of its human counterpart. We performed next-generation sequencing (NGS) and gene expression microarray profiles to explore the genomic and transcriptional backgrounds of the differential MOC line phenotypes, as well as, the cross-species relevance of the model. Here we describe the comparative analysis of NGS (www.ncbi.nlm.nih.gov/biosample?LinkName=bioproject_biosample_all&from_uid=247825) and expression microarray (www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE50041) data from the MOC lines and corresponding human data, as described in our recent publication [1].


Cancer Research | 2016

Abstract 1863: Inactivation of RASA1 promotes melanoma tumorigenesis via R-Ras activation

Kristen S. Hill; Hyeran Sung; Krishna L. Kanchi; Jane L. Messina; Ji-Hyun Lee; Young-Chul Kim; Li Ding; Richard Wilson; Jeffrey S. Weber; Minjung Kim

Hyperactivation of the Ras/Raf/Mitogen-activated protein kinase (MAPK) pathway has been commonly observed in melanoma via frequent activating mutations in NRAS and BRAF. Novel mutations in other components of this pathway such as MEK1, MEK2, MAP3K5, and MAP3K9 have been reported recently by high-throughput sequencing efforts. In addition, Ras GTPase activating proteins (RasGAPs) that negatively regulate Ras, such as NF1 (neurofibromatosis type 1) and RASA2, have been shown to be mutated or suppressed in melanoma. However, importance of other RasGAPs in melanoma has not been addressed. To obtain a comprehensive view of melanoma genomes, we conducted whole genome sequencing (WGS) of 15 metastatic melanomas and matched normal PBMC genomes from 13 melanoma patients. All melanoma genomes from these 13 patients contained at least one mutation in genes of Ras-Raf-MAPK pathway (MAPK1, MAP3K1, MAP4K2, MAP3K14, NRAS, and BRAF). In addition, we identified two novel, clustered somatic missense mutations (p.Tyr472His and p.Leu481Phe) in RASA1 (RAS p21 protein activator 1, p120RasGAP). In this study, we addressed functional roles of RASA1 in melanoma tumorigenesis. The RNAi-mediated down-regulation of RASA1 promoted, while ectopic expression of wild type RASA1 decreased, anchorage-independent colony formation, tumor growth, and RAS activation. Interestingly, RASA1 Y472H mutant enhanced soft agar colony formation and tumor growth, while RASA1 L481F mutant lost its tumor suppressive activity. Mechanistically, RASA1 required RasGAP activity to suppress colony formation and showed higher activity toward R-Ras (related RAS viral (r-ras) oncogene homolog) isoform among the Ras superfamily of small GTPases. Moreover, RASA1 consistently suppressed Ral-A among Ras downstream effectors. Reduced R-Ras or Ral-A expression via siRNAs suppressed anchorage-independent growth induced by RASA1 loss. Interestingly, RASA1 expression was frequently down-regulated in metastatic melanoma samples (11.4% (4/35) of lymph node metastasis and 3.4% (1/29) of distal metastases) compared to primary melanomas (33.3% (21/63)) and dysplastic nevi (44.1% (15/34)). We also observed significantly shorter overall survival of melanoma patients with BRAF mutations when RASA1 mRNA expression is low, which may be explained by possible cooperative interactions between activation of BRAF/MAPK/ERK and RASA1/R-Ras/Ral-A pathways. Taken together, these data support that RASA1 is a novel melanoma tumor suppressor that is inactivated by suppressed expression or by mutation. Citation Format: Kristen S. Hill, Hyeran Sung, Krishna L. Kanchi, Jane L. Messina, Ji-Hyun Lee, Youngchul Kim, Li Ding, Richard K. Wilson, Jeffrey S. Weber, Minjung Kim. Inactivation of RASA1 promotes melanoma tumorigenesis via R-Ras activation. [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 1863.


Cancer Research | 2015

Abstract A05: Identification of RASA1 as a novel melanoma tumor suppressor gene

Hyeran Sung; Krishna L. Kanchi; Jane L. Messina; Ji-Hyun Lee; Li Ding; Richard Wilson; Jeffrey S. Weber; Minjung Kim

Hyperactivation of the Ras-Raf-Mitogen-activated protein kinase (MAPK) pathway via frequent activating mutations in NRAS and BRAF has been commonly observed in melanoma. Novel mutations in other components of this pathway such as MEK1, MEK2, MAP3K5, and MAP3K9 have been reported recently by high-throughput sequencing efforts. In addition, NF1 (neurofibromatosis type 1), one of the Ras GTPase activating proteins (RasGAPs), has been shown to be mutated or suppressed in melanoma. However, importance of other RasGAPs has not been addressed in melanoma. To obtain a comprehensive view of melanoma genomes, we conducted whole genome sequencing (WGS) of 15 metastatic melanomas and matched normal PBMC genomes from 13 melanoma patients. All melanoma genomes from these 13 patients contained at least one mutation in genes of Ras-Raf-MAPK pathway (MAPK1, MAP3K1, MAP4K2, MAP3K14, NRAS, and BRAF). In addition, we identified two novel, clustered somatic missense mutations (p.Tyr472His and p.Leu481Phe) in RASA1 (RAS p21 protein activator 1, p120RasGAP) that negatively regulates Ras by catalyzing the hydrolysis of active Ras-GTP to inactive Ras-GDP. In this study, we addressed functional roles of RASA1 in melanoma tumorigenesis. Ectopic expression of wild-type RASA1 in human melanoma cell lines SKMEL28 and WM983C (with BRAF V600E) decreased, while RASA1 Y472H mutant enhanced soft agar colony formation, tumor growth, and Ras activity. The RASA1 L481F mutant lost its tumor suppressive activity. The siRNA- or shRNA-mediated knockdown of RASA1 promoted soft agar colony formation, tumor growth, and RAS activation in human melanoma cell lines IGR1 and KML1 (with BRAFV600E). Mechanistically, RASA1 required RasGAP activity to suppress colony formation and showed higher activity toward R-Ras isoform among Ras superfamily of small GTPases. To determine RASA1 level in melanocytic lesions, immunohistochemical analysis of RASA1 on human melanoma tissue microarray (TMA) was performed. RASA1 expression was frequently down-regulated in metastatic melanoma samples (11.4% (4/35) of lymph node metastasis and 3.4% (1/29) of distal metastases) compared to primary melanomas (33.3% (21/63)) and dysplastic nevi (44.1% (15/34)). Thus, these data support that RASA1 is a novel melanoma tumor suppressor that is inactivated by loss of expression or by mutation. Citation Format: Hyeran Sung, Krishna L. Kanchi, Jane Messina, Ji-Hyun Lee, Li Ding, Richard K. Wilson, Jeffrey S. Weber, Minjung Kim. Identification of RASA1 as a novel melanoma tumor suppressor gene. [abstract]. In: Proceedings of the AACR Special Conference on Advances in Melanoma: From Biology to Therapy; Sep 20-23, 2014; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(14 Suppl):Abstract nr A05.

Collaboration


Dive into the Krishna L. Kanchi's collaboration.

Top Co-Authors

Avatar

Richard Wilson

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Li Ding

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Christopher A. Miller

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Elaine R. Mardis

Nationwide Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Robert S. Fulton

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Daniel C. Koboldt

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Jane L. Messina

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Michael C. Wendl

Washington University in St. Louis

View shared research outputs
Researchain Logo
Decentralizing Knowledge