Network


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

Hotspot


Dive into the research topics where Kasthuri Kannan is active.

Publication


Featured researches published by Kasthuri Kannan.


The New England Journal of Medicine | 2014

Genetic Basis for Clinical Response to CTLA-4 Blockade in Melanoma

Alexandra Snyder; Vladimir Makarov; Taha Merghoub; Jianda Yuan; Jesse M. Zaretsky; Alexis Desrichard; Logan A. Walsh; Michael A. Postow; Phillip Wong; Teresa S. Ho; Travis J. Hollmann; Cameron Bruggeman; Kasthuri Kannan; Yanyun Li; Ceyhan Elipenahli; Cailian Liu; Christopher T. Harbison; Lisu Wang; Antoni Ribas; Jedd D. Wolchok; Timothy A. Chan

BACKGROUND Immune checkpoint inhibitors are effective cancer treatments, but molecular determinants of clinical benefit are unknown. Ipilimumab and tremelimumab are antibodies against cytotoxic T-lymphocyte antigen 4 (CTLA-4). Anti-CTLA-4 treatment prolongs overall survival in patients with melanoma. CTLA-4 blockade activates T cells and enables them to destroy tumor cells. METHODS We obtained tumor tissue from patients with melanoma who were treated with ipilimumab or tremelimumab. Whole-exome sequencing was performed on tumors and matched blood samples. Somatic mutations and candidate neoantigens generated from these mutations were characterized. Neoantigen peptides were tested for the ability to activate lymphocytes from ipilimumab-treated patients. RESULTS Malignant melanoma exomes from 64 patients treated with CTLA-4 blockade were characterized with the use of massively parallel sequencing. A discovery set consisted of 11 patients who derived a long-term clinical benefit and 14 patients who derived a minimal benefit or no benefit. Mutational load was associated with the degree of clinical benefit (P=0.01) but alone was not sufficient to predict benefit. Using genomewide somatic neoepitope analysis and patient-specific HLA typing, we identified candidate tumor neoantigens for each patient. We elucidated a neoantigen landscape that is specifically present in tumors with a strong response to CTLA-4 blockade. We validated this signature in a second set of 39 patients with melanoma who were treated with anti-CTLA-4 antibodies. Predicted neoantigens activated T cells from the patients treated with ipilimumab. CONCLUSIONS These findings define a genetic basis for benefit from CTLA-4 blockade in melanoma and provide a rationale for examining exomes of patients for whom anti-CTLA-4 agents are being considered. (Funded by the Frederick Adler Fund and others.).


The Journal of Clinical Endocrinology and Metabolism | 2013

Genomic Dissection of Hurthle Cell Carcinoma Reveals a Unique Class of Thyroid Malignancy

Ian Ganly; Julio Ricarte Filho; Stephanie Eng; Ronald Ghossein; Luc G. Morris; Yupu Liang; Nicholas D. Socci; Kasthuri Kannan; Qianxing Mo; James A. Fagin; Timothy A. Chan

CONTEXT Hurthle cell cancer (HCC) is an understudied cancer with poor prognosis. OBJECTIVE Our objective was to elucidate the genomic foundations of HCC. DESIGN AND SETTING We conducted a large-scale integrated analysis of mutations, gene expression profiles, and copy number alterations in HCC at a single tertiary-care cancer institution. METHODS Mass spectrometry-based genotyping was used to interrogate hot spot point mutations in the most common thyroid oncogenes: BRAF, RET, NRAS, HRAS, KRAS, PIK3CA, MAP2K1, and AKT1. In addition, common oncogenic fusions of RET and NTRK1 as well as PAX8/PPARγ and AKAP9-BRAF were also assessed by RT-PCR. Global copy number changes and gene expression profiles were determined in the same tumor set as the mutational analyses. RESULTS We report that the mutational, transcriptional, and copy number profiles of HCC were distinct from those of papillary thyroid cancer and follicular thyroid cancer, indicating HCC to be a unique type of thyroid malignancy. Unsupervised hierarchical clustering of gene expression showed the 3 groups of Hurthle tumors (Hurthle cell adenoma [HA], minimally invasive Hurthle cell carcinoma [HMIN], and widely invasive Hurthle cell carcinoma [HWIDE] clustered separately with a marked difference between HWIDE and HA. Global copy number analysis also indicated distinct subgroups of tumors that may arise as HWIDE and HMIN. Molecular pathways that differentiate HA from HWIDE included the PIK3CA-Akt-mTOR and Wnt/β-catenin pathways, potentially providing a rationale for new targets for this type of malignancy. CONCLUSIONS Our data provide evidence that HCC may be a unique thyroid cancer distinct from papillary and follicular thyroid cancer.


Nature Communications | 2018

Atrx inactivation drives disease-defining phenotypes in glioma cells of origin through global epigenomic remodeling

Carla Danussi; Promita Bose; Prasanna Parthasarathy; Pedro Silberman; John S. Van Arnam; Mark Vitucci; Oliver Tang; Adriana Heguy; Yuxiang Wang; Timothy A. Chan; Gregory J. Riggins; Erik P. Sulman; Frederick F. Lang; Chad J. Creighton; Benjamin Deneen; C. Ryan Miller; David J. Picketts; Kasthuri Kannan; Jason T. Huse

Mutational inactivation of the SWI/SNF chromatin regulator ATRX occurs frequently in gliomas, the most common primary brain tumors. Whether and how ATRX deficiency promotes oncogenesis by epigenomic dysregulation remains unclear, despite its recent implication in both genomic instability and telomere dysfunction. Here we report that Atrx loss recapitulates characteristic disease phenotypes and molecular features in putative glioma cells of origin, inducing cellular motility although also shifting differentiation state and potential toward an astrocytic rather than neuronal histiogenic profile. Moreover, Atrx deficiency drives widespread shifts in chromatin accessibility, histone composition, and transcription in a distribution almost entirely restricted to genomic sites normally bound by the protein. Finally, direct gene targets of Atrx that mediate specific Atrx-deficient phenotypes in vitro exhibit similarly selective misexpression in ATRX-mutant human gliomas. These findings demonstrate that ATRX deficiency and its epigenomic sequelae are sufficient to induce disease-defining oncogenic phenotypes in appropriate cellular and molecular contexts.ATRX inactivation frequently occurs in glioma. Here, the authors explore the role of ATRX inactivation in oncogenesis, highlighting ATRX deficiency driven epigenomic changes that influence the expression of genes crucial to the oncogenic phenotype.


Journal of Investigative Dermatology | 2017

Role of Dysregulated Cytokine Signaling and Bacterial Triggers in the Pathogenesis of Cutaneous T-Cell Lymphoma

Melania H. Fanok; Amy Sun; Laura K. Fogli; Vijay Narendran; Miriam Eckstein; Kasthuri Kannan; Igor Dolgalev; Charalampos Lazaris; Adriana Heguy; Mary E. Laird; Mark S. Sundrud; Cynthia Liu; Jeff Kutok; Rodrigo S. Lacruz; Jo Ann Latkowski; Iannis Aifantis; Niels Ødum; Kenneth B. Hymes; Swati Goel; Sergei B. Koralov

Cutaneous T-cell lymphoma is a heterogeneous group of lymphomas characterized by the accumulation of malignant T cells in the skin. The molecular and cellular etiology of this malignancy remains enigmatic, and what role antigenic stimulation plays in the initiation and/or progression of the disease remains to be elucidated. Deep sequencing of the tumor genome showed a highly heterogeneous landscape of genetic perturbations, and transcriptome analysis of transformed T cells further highlighted the heterogeneity of this disease. Nonetheless, using data harvested from high-throughput transcriptional profiling allowed us to develop a reliable signature of this malignancy. Focusing on a key cytokine signaling pathway previously implicated in cutaneous T-cell lymphoma pathogenesis, JAK/STAT signaling, we used conditional gene targeting to develop a fully penetrant small animal model of this disease that recapitulates many key features of mycosis fungoides, a common variant of cutaneous T-cell lymphoma. Using this mouse model, we show that T-cell receptor engagement is critical for malignant transformation of the T lymphocytes and that progression of the disease is dependent on microbiota.


Clinical Cancer Research | 2016

Abstract 11: Advancing methylation profiling in neuropathology: Diagnosis and clinical management

Kasthuri Kannan; Aristotelis Tsirigos; Jonathan Serrano; Lynn Ann Forrester; Arline Faustin; Cheddhi Thomas; David Capper; Volker Hovestadt; Stefan M. Pfister; David T. W. Jones; Martin Sill; Daniel Schrimpf; Andreas von Deimling; Adriana Heguy; Sharon Gardner; Jeffrey C. Allen; Cyrus Hedvat; David Zagzag; Matija Snuderl; Matthias A. Karajannis

Introduction: Although molecular profiling is increasingly being applied to improve subgroup classification and to provide novel prognostic and predictive biomarkers, clinical neuropathology practice is largely based on morphology and immunohistochemistry. Current molecular methods play only a small role in determining the diagnosis itself. Methods: For molecular subclassification of tumors at NYU neuropathology and to improve diagnostic accuracy, we introduced genome-wide methylation profiling through Illumina Infinium HumanMethylation 450k array that can detect methylation marks from the DNA extracted from formalin-fixed paraffin embedded tissues. To this effort, an in-house pipeline was established in-house pipeline, which includes morphologic review, sample preparation, molecular profiling and bioinformatics analysis. We compared the methylation profiles to a reference cohort of 2150 cases from 77 tumor entities previously profiled and analyzed at German Cancer Research Center using a random forest algorithm and customized bioinformatics packages, which were shared between our institutions. Selected copy number variants (CNV) and mutations were confirmed by Fluorescence in situ Hybridization (FISH) or sequencing, and mutation specific immunohistochemistry, respectively. Results: We profiled 60 difficult in-house or consult adult and pediatric brain tumors where diagnosis, grade and/or molecular subtype were not conclusive by morphology, immunohistochemistry or standard molecular studies alone. There was 100% concordance with concurrently performed molecular tests such as 1p/19q, EGFR/BRAF CNV, MGMT promoter methylation or IDH1 status testing when these tests were performed for clinical care. Methylation profiling provided additional, relevant information in 30 of 60 (50%) cases, leading to a change of diagnosis in 9 (15%), clarification of the diagnosis in 7 (12%) cases, and further molecular subgroup refinement in 14 (23%) of cases, helping to direct further molecular testing and clinical management. Conclusion: The 450k methylation array platform represents a cost-efficient method to obtain molecular profiles of brain tumors to identify biologically relevant diagnostic subgroups, thereby improving diagnostic accuracy, and helping inform appropriate clinical management decisions. Citation Format: Kasthuri S. Kannan, Aristotelis Tsirigos, Jonathan Serrano, Lynn Ann Forrester, Arline Faustin, Cheddhi Thomas, David Capper, Volker Hovestadt, Stefan M. Pfister, David T. W Jones, Martin Sill, Daniel Schrimpf, Andreas von Deimling, Adriana Heguy, Sharon L. Gardner, Jeffrey Allen, Cyrus Hedvat, David Zagzag, Matija Snuderl, Matthias A. Karajannis. Advancing methylation profiling in neuropathology: Diagnosis and clinical management. [abstract]. In: Proceedings of the AACR Precision Medicine Series: Integrating Clinical Genomics and Cancer Therapy; Jun 13-16, 2015; Salt Lake City, UT. Philadelphia (PA): AACR; Clin Cancer Res 2016;22(1_Suppl):Abstract nr 11.


npj Genomic Medicine | 2018

Cardiac arrhythmia and neuroexcitability gene variants in resected brain tissue from patients with sudden unexpected death in epilepsy (SUDEP)

Daniel Friedman; Kasthuri Kannan; Arline Faustin; Seema Shroff; Cheddhi Thomas; Adriana Heguy; Jonathan Serrano; Matija Snuderl; Orrin Devinsky

Sudden unexpected death in epilepsy (SUDEP) is the leading cause of epilepsy-related mortality in young adults. The exact mechanisms are unknown but death often follows a generalized tonic–clonic seizure. Proposed mechanisms include seizure-related respiratory, cardiac, autonomic, and arousal dysfunction. Genetic drivers underlying SUDEP risk are largely unknown. To identify potential SUDEP risk genes, we compared whole-exome sequences (WES) derived from formalin-fixed paraffin embedded surgical brain specimens of eight epilepsy patients who died from SUDEP with seven living controls matched for age at surgery, sex, year of surgery and lobe of resection. We compared identified variants from both groups filtering known polymorphisms from publicly available data as well as scanned for epilepsy and candidate SUDEP genes. In the SUDEP cohort, we identified mutually exclusive variants in genes involved in µ-opiod signaling, gamma-aminobutyric acid (GABA) and glutamate-mediated synaptic signaling, including ARRB2, ITPR1, GABRR2, SSTR5, GRIK1, CTNAP2, GRM8, GNAI2 and GRIK5. In SUDEP patients we also identified variants in genes associated with cardiac arrhythmia, including KCNMB1, KCNIP1, DPP6, JUP, F2, and TUBA3D, which were not present in living epilepsy controls. Our data shows that genomic analysis of brain tissue resected for seizure control can identify potential genetic biomarkers of SUDEP risk.Epilepsy: Spotting genetic drivers of sudden deathGene variants associated with abnormal heart rhythm and neuronal excitability may increase the risk of Sudden Unexpected Death in Epilepsy (SUDEP). SUDEP is the most common cause of death directly related to epilepsy, but little is known about the risk factors and mechanisms through which seizures can lead to death. Daniel Friedman, Orrin Devinsky and colleagues at New York University Langone Medical Center, US, compared whole-exome sequences from brain tissue belonging to eight epilepsy patients who died from SUDEP and seven matched living controls who had brain tissue removed for epilepsy treatment. In the SUDEP cases they identified 13 rare gene variants involved in cardiac arrhythmia and excitatory neurotransmission as potential genetic biomarkers of SUDEP risk. Further understanding the genetic contribution to epilepsy-related mortality will help develop effective preventive strategies.


bioRxiv | 2018

G-quadruplex DNA drives genomic instability and represents a targetable molecular abnormality in ATRX-deficient malignant glioma

Yuxiang Wang; Jie Yang; Wei H. Wu; Rachna Shah; Carla Danussi; Gregory J. Riggins; Kasthuri Kannan; Erik P. Sulman; Timothy A. Chan; Jason T. Huse

Mutational inactivation of ATRX (α-thalassemia mental retardation X-linked) represents a defining molecular alteration in large subsets of malignant glioma. Yet the pathogenic consequences of ATRX deficiency remain unclear, as do tractable mechanisms for its therapeutic targeting. Here we report that ATRX loss in isogenic glioma model systems induces replication stress and DNA damage by way of G-quadruplex (G4) DNA secondary structure. Moreover, these effects are associated with the acquisition of disease-relevant copy number alterations over time. We then demonstrate, both in vitro and in vivo, that ATRX deficiency selectively enhances DNA damage and cell death following chemical G4 stabilization. Finally, we show that G4 stabilization synergizes with other DNA-damaging therapies, including ionizing radiation, in the ATRX-deficient context. Our findings reveal novel pathogenic mechanisms driven by ATRX deficiency in glioma, while also pointing to tangible strategies for drug development.


Nature Communications | 2018

Recurrent homozygous deletion of DROSHA and microduplication of PDE4DIP in pineoblastoma

Matija Snuderl; Kasthuri Kannan; Elke Pfaff; Shiyang Wang; James M. Stafford; Jonathan Serrano; Adriana Heguy; Karina Ray; Arline Faustin; Olga Aminova; Igor Dolgalev; Stacie Stapleton; David Zagzag; Luis Chiriboga; Sharon Gardner; Jeffrey H. Wisoff; John G. Golfinos; David Capper; Volker Hovestadt; Marc K. Rosenblum; Dimitris G. Placantonakis; Sarah LeBoeuf; Thales Papagiannakopoulos; Lukas Chavez; Sama Ahsan; Charles G. Eberhart; Stefan M. Pfister; David T. W. Jones; Matthias A. Karajannis

Pineoblastoma is a rare and highly aggressive brain cancer of childhood, histologically belonging to the spectrum of primitive neuroectodermal tumors. Patients with germline mutations in DICER1, a ribonuclease involved in microRNA processing, have increased risk of pineoblastoma, but genetic drivers of sporadic pineoblastoma remain unknown. Here, we analyzed pediatric and adult pineoblastoma samples (n = 23) using a combination of genome-wide DNA methylation profiling and whole-exome sequencing or whole-genome sequencing. Pediatric and adult pineoblastomas showed distinct methylation profiles, the latter clustering with lower-grade pineal tumors and normal pineal gland. Recurrent variants were found in genes involved in PKA- and NF-κB signaling, as well as in chromatin remodeling genes. We identified recurrent homozygous deletions of DROSHA, acting upstream of DICER1 in microRNA processing, and a novel microduplication involving chromosomal region 1q21 containing PDE4DIP (myomegalin), comprising the ancient DUF1220 protein domain. Expresion of PDE4DIP and DUF1220 proteins was present exclusively in pineoblastoma with PDE4DIP gain.Pineoblastoma is a highly aggressive and rare childhood brain cancer, and the genetic drivers of sporadic pineoblastoma are unknown. Here, the authors genomically interrogated pediatric and adult pineoblastomas and found novel variants including recurrent homozygous deletions of DROSHA.


bioRxiv | 2016

Why do mutant allele frequencies in oncogenes peak around .40 and rapidly decrease

Kasthuri Kannan; Adriana Heguy

The mutant allele frequencies in oncogenes peak around .40 and rapidly decrease. In this article, we explain why this is the case. Invoking a key result from mathematical analysis in our model, namely, the inverse function theorem, we estimate the selection coefficients of the mutant alleles as a function of germline allele frequencies. Under complete dominance of oncogenic mutations, this selection function is expected to be linearly correlated with the distribution of the mutant alleles. We demonstrate that this is the case by investigating the allele frequencies of mutations in oncogenes across various cancer types, validating our model for mean effective selection. Consistent with the population genetics model of fitness, the selection function fits a gamma-distribution curve that accurately describes the trend of the mutant allele frequencies. While existing equations for selection explain evolution at low allele frequencies, our equations are general formulas for natural selection under complete dominance operating at all frequencies. We show that selection exhibits linear behaviour at all times, favouring dominant alleles with respect to the change in recessive allele frequencies. Also, these equations show, selection behaves like power law against the recessive alleles at low dominant allele frequencies.


Cancer Research | 2016

Abstract P1-06-06: Mammary stem cell modulation of wildtype and Trp53 null stem cells by CAPE (caffeic acid phenethyl ester), a potential therapeutic agent

Coral Omene; Manan Patel; Kasthuri Kannan; Adriana Heguy; Mary Helen Barcellos-Hoff

CAPE is the major active component of propolis, a widely available, non-toxic, honeybee natural product with anti-inflammatory, antioxidant, and antitumor properties. We have previously shown that CAPE inhibits growth of breast cancer cells and the tumorigenic potential of breast cancer stem cells. We have identified inhibition of histone deacetylase (HDAC) as one mechanism of action, which suggests that it mediates its effects through epigenetic modifications. We postulated that CAPE may be useful in chemoprevention for women at high risk for triple-negative breast cancers since the cell-of-origin hypothesis states that these cancers likely arise from transformation of mammary stem or progenitor cells, whose self-renewal is maintained via epigenetic states. We tested the effect of CAPE on wildtype (WT) and Trp53 null mammary stem cell (MaSC) self-renewal from BALB/c mice cultured as mammospheres (MMS). Primary mammary epithelial cells were cultured as MMS for 7 days, dissociated into single cells, re-cultured in the presence of CAPE for 7 days and passaged in secondary and tertiary passages without CAPE. MMS frequency and differentiation potential was analyzed using immunofluorescence detection of luminal marker, cytokeratin 18, basal marker, cytokeratin 14, and progesterone receptor (PR). Chromatin states were identified using ATAC-seq and open chromatin areas unique to CAPE treated murine MMS were used for pathway analysis performed by Ingenuity Pathway Analysis, Gene Set Enrichment Analysis and confirmed by Integrative Genome Viewer. CAPE treatment resulted in a dose dependent decrease in both WT and p53 null mammosphere forming efficiency that persisted in secondary and tertiary passages, suggesting reduced self-renewal. CAPE treatment also shifted differentiation from predominantly basal K14 to luminal K18-positive in both WT and p53 null MMS and increased PR expression in WT MMS. ATAC-seq of CAPE treated WT MMS showed significant pathway enrichment for p53 signaling, SOX2 signaling, and enrichment of open chromatin for several genes including the SMARCA4 gene, which regulates transcription of genes involved in stem cell renewal. ATAC-seq of CAPE treated Trp53 null MMS showed that genes defining early and late response to estrogen were particularly important. Significant canonical pathways included Aryl hydrocarbon receptor signaling, whose upregulation results in inhibition of self renewal and has been targeted as a potential drug target for estrogen receptor negative breast cancer. The integrin signaling pathway was also highly enriched. These data suggest that CAPE both inhibits MaSC self-renewal and shifts the lineage commitment to a luminal, ER + lineage. ATAC-seq demonstrated genomic effects that are important in differentiation, SC renewal and adhesion. These data suggest that CAPE may have an effect on lineage commitment in support of our chemoprevention strategy to reduce triple-negative breast cancer. Citation Format: Omene C, Patel M, Kannan K, Heguy A, Barcellos-Hoff MH. Mammary stem cell modulation of wildtype and Trp53 null stem cells by CAPE (caffeic acid phenethyl ester), a potential therapeutic agent. [abstract]. In: Proceedings of the Thirty-Eighth Annual CTRC-AACR San Antonio Breast Cancer Symposium: 2015 Dec 8-12; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2016;76(4 Suppl):Abstract nr P1-06-06.

Collaboration


Dive into the Kasthuri Kannan's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Timothy A. Chan

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

David T. W. Jones

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Stefan M. Pfister

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Volker Hovestadt

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Matthias A. Karajannis

Memorial Sloan Kettering Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge