Network


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

Hotspot


Dive into the research topics where Vinod P. Balachandran is active.

Publication


Featured researches published by Vinod P. Balachandran.


Nature Medicine | 2011

Imatinib potentiates antitumor T cell responses in gastrointestinal stromal tumor through the inhibition of Ido

Vinod P. Balachandran; Michael J. Cavnar; Shan Zeng; Zubin M. Bamboat; Lee M. Ocuin; Hebroon Obaid; Eric C. Sorenson; Rachel Popow; Charlotte Ariyan; Ferdinand Rossi; Peter Besmer; Tianhua Guo; Cristina R. Antonescu; Takahiro Taguchi; Jianda Yuan; Jedd D. Wolchok; James P. Allison; Ronald P. DeMatteo

Imatinib mesylate targets mutated KIT oncoproteins in gastrointestinal stromal tumor (GIST) and produces a clinical response in 80% of patients. The mechanism is believed to depend predominantly on the inhibition of KIT-driven signals for tumor-cell survival and proliferation. Using a mouse model of spontaneous GIST, we found that the immune system contributes substantially to the antitumor effects of imatinib. Imatinib therapy activated CD8+ T cells and induced regulatory T cell (Treg cell) apoptosis within the tumor by reducing tumor-cell expression of the immunosuppressive enzyme indoleamine 2,3-dioxygenase (Ido). Concurrent immunotherapy augmented the efficacy of imatinib in mouse GIST. In freshly obtained human GIST specimens, the T cell profile correlated with imatinib sensitivity and IDO expression. Thus, T cells are crucial to the antitumor effects of imatinib in GIST, and concomitant immunotherapy may further improve outcomes in human cancers treated with targeted agents.


Hepatology | 2010

Toll‐like receptor 9 inhibition confers protection from liver ischemia–reperfusion injury

Zubin M. Bamboat; Vinod P. Balachandran; Lee M. Ocuin; Hebroon Obaid; George Plitas; Ronald P. DeMatteo

Endogenous ligands such as high‐mobility group box 1 (HMGB1) and nucleic acids are released by dying cells and bind Toll‐like receptors (TLRs). Because TLR9 sits at the interface of microbial and sterile inflammation by detecting both bacterial and endogenous DNA, we investigated its role in a model of segmental liver ischemia–reperfusion (I/R) injury. Mice were subjected to 1 hour of ischemia and 12 hours of reperfusion before assessment of liver injury, cytokines, and reactive oxygen species (ROS). Wild‐type (WT) mice treated with an inhibitory cytosine‐guanosine dinucleotide (iCpG) sequence and TLR9−/− mice had markedly reduced serum alanine aminotransferase (ALT) and inflammatory cytokines after liver I/R. Liver damage was mediated by bone marrow–derived cells because WT mice transplanted with TLR9−/− bone marrow were protected from hepatic I/R injury. Injury in WT mice partly depended on TLR9 signaling in neutrophils, which enhanced production of ROS, interleukin‐6 (IL‐6), and tumor necrosis factor (TNF). In vitro, DNA released from necrotic hepatocytes increased liver nonparenchymal cell (NPC) and neutrophil cytokine secretion through a TLR9‐dependent mechanism. Inhibition of both TLR9 and HMGB1 caused maximal inflammatory cytokine suppression in neutrophil cultures and conferred even greater protection from I/R injury in vivo. Conclusion: TLR9 serves as an endogenous sensor of tissue necrosis that exacerbates the innate immune response during liver I/R. Combined blockade of TLR9 and HMGB1 represents a clinically relevant, novel approach to limiting I/R injury. (HEPATOLOGY 2009.)


Journal of Clinical Investigation | 2010

Conventional DCs reduce liver ischemia/reperfusion injury in mice via IL-10 secretion

Zubin M. Bamboat; Lee M. Ocuin; Vinod P. Balachandran; Hebroon Obaid; George Plitas; Ronald P. DeMatteo

TLRs are recognized as promoters of tissue damage, even in the absence of pathogens. TLR binding to damage-associated molecular patterns (DAMPs) released by injured host cells unleashes an inflammatory cascade that amplifies tissue destruction. However, whether TLRs possess the reciprocal ability to curtail the extent of sterile inflammation is uncertain. Here, we investigated this possibility in mice by studying the role of conventional DCs (cDCs) in liver ischemia/reperfusion (I/R) injury, a model of sterile inflammation. Targeted depletion of mouse cDCs increased liver injury after I/R, as assessed by serum alanine aminotransferase and histologic analysis. In vitro, we identified hepatocyte DNA as an endogenous ligand to TLR9 that promoted cDCs to secrete IL-10. In vivo, cDC production of IL-10 required TLR9 and reduced liver injury. In addition, we found that inflammatory monocytes recruited to the liver via chemokine receptor 2 were downstream targets of cDC IL-10. IL-10 from cDCs reduced production of TNF, IL-6, and ROS by inflammatory monocytes. Our results implicate inflammatory monocytes as mediators of liver I/R injury and reveal that cDCs respond to DAMPS during sterile inflammation, providing the host with protection from progressive tissue damage.


Lancet Oncology | 2015

Nomograms in oncology: more than meets the eye

Vinod P. Balachandran; Mithat Gonen; J. Joshua Smith; Ronald P. DeMatteo

Nomograms are widely used as prognostic devices in oncology and medicine. With the ability to generate an individual probability of a clinical event by integrating diverse prognostic and determinant variables, nomograms meet our desire for biologically and clinically integrated models and fulfill our drive towards personalised medicine. Rapid computation through user-friendly digital interfaces, together with increased accuracy, and more easily understood prognoses compared with conventional staging, allow for seamless incorporation of nomogram-derived prognosis to aid clinical decision making. This has led to the appearance of many nomograms on the internet and in medical journals, and an increase in nomogram use by patients and physicians alike. However, the statistical foundations of nomogram construction, their precise interpretation, and evidence supporting their use are generally misunderstood. This issue is leading to an under-appreciation of the inherent uncertainties regarding nomogram use. We provide a systematic, practical approach to evaluating and comprehending nomogram-derived prognoses, with particular emphasis on clarifying common misconceptions and highlighting limitations.


Journal of Experimental Medicine | 2013

KIT oncogene inhibition drives intratumoral macrophage M2 polarization

Michael J. Cavnar; Shan Zeng; Teresa S. Kim; Eric C. Sorenson; Lee M. Ocuin; Vinod P. Balachandran; Adrian M. Seifert; Jonathan B. Greer; Rachel Popow; Megan H. Crawley; Noah A. Cohen; Benjamin L. Green; Ferdinand Rossi; Peter Besmer; Cristina R. Antonescu; Ronald P. DeMatteo

Imatinib reduces tumor cell KIT signaling and causes tumor cell apoptosis, which drives TAMs to shift from M1- to M2-like in mouse and human GIST.


Journal of Leukocyte Biology | 2011

Neutrophil IL-10 suppresses peritoneal inflammatory monocytes during polymicrobial sepsis

Lee M. Ocuin; Zubin M. Bamboat; Vinod P. Balachandran; Michael J. Cavnar; Hebroon Obaid; George Plitas; Ronald P. DeMatteo

Septic peritonitis remains a major cause of death. Neutrophils and inflammatory monocytes are principal components of the innate immune system and are essential for defense against a range of microbial pathogens. Their role and interaction in polymicrobial sepsis have not been defined clearly. Using a murine model of CLP to induce moderate sepsis, we found that neutrophil depletion did not alter survival, whereas depletion of neutrophils and inflammatory monocytes markedly reduced survival. After neutrophil depletion, inflammatory monocytes had greater phagocytic capacity and oxidative burst, and increased expression of costimulatory molecules, TNF, and iNOS. Notably, peritoneal neutrophils produced IL‐10 following CLP. Adoptive i.p. transfer of WT but not IL‐10−/− neutrophils into septic mice reduced monocyte expression of TNF. In vitro experiments confirmed that monocyte suppression was mediated by neutrophil‐derived IL‐10. Thus, during septic peritonitis, neutrophils suppress peritoneal inflammatory monocytes through IL‐10 and are dispensable for survival.


Nature | 2017

A neoantigen fitness model predicts tumour response to checkpoint blockade immunotherapy

Marta Łuksza; Nadeem Riaz; Vladimir Makarov; Vinod P. Balachandran; Matthew D. Hellmann; Alexander Solovyov; Naiyer A. Rizvi; Taha Merghoub; Arnold J. Levine; Timothy A. Chan; Jedd D. Wolchok; Benjamin D. Greenbaum

Checkpoint blockade immunotherapies enable the host immune system to recognize and destroy tumour cells. Their clinical activity has been correlated with activated T-cell recognition of neoantigens, which are tumour-specific, mutated peptides presented on the surface of cancer cells. Here we present a fitness model for tumours based on immune interactions of neoantigens that predicts response to immunotherapy. Two main factors determine neoantigen fitness: the likelihood of neoantigen presentation by the major histocompatibility complex (MHC) and subsequent recognition by T cells. We estimate these components using the relative MHC binding affinity of each neoantigen to its wild type and a nonlinear dependence on sequence similarity of neoantigens to known antigens. To describe the evolution of a heterogeneous tumour, we evaluate its fitness as a weighted effect of dominant neoantigens in the subclones of the tumour. Our model predicts survival in anti-CTLA-4-treated patients with melanoma and anti-PD-1-treated patients with lung cancer. Importantly, low-fitness neoantigens identified by our method may be leveraged for developing novel immunotherapies. By using an immune fitness model to study immunotherapy, we reveal broad similarities between the evolution of tumours and rapidly evolving pathogens.


Clinical Cancer Research | 2017

Stratification of Pancreatic Ductal Adenocarcinoma: Combinatorial Genetic, Stromal, and Immunologic Markers

Erik S. Knudsen; Paris Vail; Uthra Balaji; Hoai Ngo; Ihab W. Botros; Vladimir Makarov; Nadeem Riaz; Vinod P. Balachandran; Steven D. Leach; Debrah Thompson; Timothy A. Chan; Agnieszka K. Witkiewicz

Purpose: Pancreatic ductal adenocarcinoma (PDAC) is associated with an immunosuppressive milieu that supports immune system evasion and disease progression. Here, we interrogated genetic, stromal, and immunologic features of PDAC to delineate impact on prognosis and means to more effectively employ immunotherapy. Experimental Design: A cohort of 109 PDAC cases annotated for overall survival was utilized as a primary discovery cohort. Gene expression analysis defined immunologic subtypes of PDAC that were confirmed in the Cancer Genome Atlas dataset. Stromal and metabolic characteristics of PDAC cases were evaluated by histologic analysis and immunostaining. Enumeration of lymphocytes, as well as staining for CD8, FOXP3, CD68, CD163, PDL1, and CTLA4 characterized immune infiltrate. Neoantigens were determined by analysis of whole-exome sequencing data. Random-forest clustering was employed to define multimarker subtypes, with univariate and multivariate analyses interrogating prognostic significance. Results: PDAC cases exhibited distinct stromal phenotypes that were associated with prognosis, glycolytic and hypoxic biomarkers, and immune infiltrate composition. Immune infiltrate was diverse among PDAC cases and enrichment for M2 macrophages and select immune checkpoints regulators were specifically associated with survival. Composite analysis with neoantigen burden, immunologic, and stromal features defined novel subtypes of PDAC that could have bearing on sensitivity to immunologic therapy approaches. In addition, a subtype with low levels of neoantigens and minimal lymphocyte infiltrate was associated with improved overall survival. Conclusions: The mutational burden of PDAC is associated with distinct immunosuppressive mechanisms that are conditioned by the tumor stromal environment. The defined subtypes have significance for utilizing immunotherapy in the treatment of PDAC. Clin Cancer Res; 23(15); 4429–40. ©2017 AACR.


Clinical Cancer Research | 2017

Real Time Genomic Profiling of Pancreatic Ductal Adenocarcinoma: Potential Actionability and Correlation with Clinical Phenotype

Maeve Aine Lowery; Emmet Jordan; Olca Basturk; Ryan Ptashkin; Ahmet Zehir; Michael F. Berger; Tanisha Leach; Brian Herbst; Gokce Askan; Hannah Maynard; Danielle C. Glassman; Christina M. Covington; Nikolaus Schultz; Ghassan K. Abou-Alfa; James J. Harding; David S. Klimstra; Jaclyn F. Hechtman; David M. Hyman; Peter J. Allen; William R. Jarnagin; Vinod P. Balachandran; Anna M. Varghese; Mark A. Schattner; Kenneth H. Yu; Leonard Saltz; David B. Solit; Christine A. Iacobuzio-Donahue; Steven D. Leach; Eileen Mary O'Reilly

Purpose: Molecular profiling in cancer has identified potential actionable drug targets that have prompted attempts to discover clinically validated biomarkers to guide therapeutic decision-making and enrollment to clinical trials. We evaluated whether comprehensive genetic analysis of patients with pancreatic adenocarcinoma is feasible within a clinically relevant timeframe and whether such analyses provide predictive and/or prognostic information along with identification of potential targets for therapy. Experimental Design: Archival or prospectively acquired FFPE samples and matched normal DNA from N = 336 patients with pancreatic cancer were analyzed using a hybridization capture–based, next-generation sequencing assay designed to perform targeted deep sequencing of all exons and selected introns of 410 key cancer-associated genes. Demographic and treatment data were prospectively collected with the goal of correlating treatment outcomes and drug response with molecular profiles. Results: The median time from protocol consent to reporting of the genomic results was 45 days with a median time from tissue delivery of 20 days. All genetic alterations identified were stratified based upon prior evidence that the mutation is a predictive biomarker of drug response using the MSKCC OncoKB classification. Three of 225 patients (1%) received a matched therapy based upon the sequencing results. Conclusions: The practical application of molecular results to guide individual patient treatment is currently limited in patients with pancreatic adenocarcinoma. Future prospective molecular profiling efforts should seek to incorporate routine germline genetic analysis and the identification of DNA profiles that predict for clinical benefit from agents that target DNA damage repair and or immunotherapy. Clin Cancer Res; 23(20); 6094–100. ©2017 AACR.


Journal of Surgical Oncology | 2017

Recurrence patterns following irreversible electroporation for hepatic malignancies

Russell C. Langan; Debra A. Goldman; Michael I. D'Angelica; Ronald P. DeMatteo; Peter J. Allen; Vinod P. Balachandran; William R. Jarnagin; T. Peter Kingham

Irreversible electroporation (IRE) has emerged as a novel, safe ablative therapy for peri‐vascular lesions. However, there remains a paucity of data on long‐term outcomes.

Collaboration


Dive into the Vinod P. Balachandran's collaboration.

Top Co-Authors

Avatar

Ronald P. DeMatteo

Memorial Hospital of South Bend

View shared research outputs
Top Co-Authors

Avatar

Peter J. Allen

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

William R. Jarnagin

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

T. Peter Kingham

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Michael I. D'Angelica

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Michael I. D’Angelica

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

T.P. Kingham

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Amber L. Simpson

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Linda M. Pak

Memorial Sloan Kettering Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge