Network


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

Hotspot


Dive into the research topics where Samir Kharbanda is active.

Publication


Featured researches published by Samir Kharbanda.


Cancer Cell | 2010

A Hierarchy of Self-Renewing Tumor-Initiating Cell Types in Glioblastoma

Ruihuan Chen; Merry Nishimura; Stephanie M. Bumbaca; Samir Kharbanda; William F. Forrest; Ian Kasman; Joan M. Greve; Robert Soriano; Laurie L. Gilmour; Celina Sanchez Rivers; Zora Modrusan; Serban Nacu; Steve Guerrero; Kyle A. Edgar; Jeffrey Wallin; Katrin Lamszus; Manfred Westphal; Susanne Heim; C. David James; Scott R. VandenBerg; Joseph F. Costello; Scott Moorefield; Cynthia Cowdrey; Michael D. Prados; Heidi S. Phillips

The neural stem cell marker CD133 is reported to identify cells within glioblastoma (GBM) that can initiate neurosphere growth and tumor formation; however, instances of CD133(-) cells exhibiting similar properties have also been reported. Here, we show that some PTEN-deficient GBM tumors produce a series of CD133(+) and CD133(-) self-renewing tumor-initiating cell types and provide evidence that these cell types constitute a lineage hierarchy. Our results show that the capacities for self-renewal and tumor initiation in GBM need not be restricted to a uniform population of stemlike cells, but can be shared by a lineage of self-renewing cell types expressing a range of markers of forebrain lineage.


Journal of Clinical Oncology | 2011

Evidence for Sequenced Molecular Evolution of IDH1 Mutant Glioblastoma From a Distinct Cell of Origin

Albert Lai; Samir Kharbanda; Whitney B. Pope; Anh Tran; Orestes E. Solis; Franklin Peale; William F. Forrest; Kanan Pujara; Jose Carrillo; Ajay Pandita; Benjamin M. Ellingson; Chauncey W. Bowers; Robert Soriano; Nils Ole Schmidt; Sankar Mohan; William H. Yong; Somasekar Seshagiri; Zora Modrusan; Zhaoshi Jiang; Kenneth D. Aldape; Paul S. Mischel; Linda M. Liau; Cameron Escovedo; Weidong Chen; Phioanh L. Nghiemphu; C. David James; Michael D. Prados; Manfred Westphal; Katrin Lamszus; Timothy F. Cloughesy

PURPOSE Mutation in isocitrate dehydrogenase 1 (IDH1) at R132 (IDH1(R132MUT)) is frequent in low-grade diffuse gliomas and, within glioblastoma (GBM), has been proposed as a marker for GBMs that arise by transformation from lower-grade gliomas, regardless of clinical history. To determine how GBMs arising with IDH1(R132MUT) differ from other GBMs, we undertook a comprehensive comparison of patients presenting clinically with primary GBM as a function of IDH1(R132) mutation status. PATIENTS AND METHODS In all, 618 treatment-naive primary GBMs and 235 lower-grade diffuse gliomas were sequenced for IDH1(R132) and analyzed for demographic, radiographic, anatomic, histologic, genomic, epigenetic, and transcriptional characteristics. RESULTS Investigation revealed a constellation of features that distinguishes IDH1(R132MUT) GBMs from other GBMs (including frontal location and lesser extent of contrast enhancement and necrosis), relates them to lower-grade IDH1(R132MUT) gliomas, and supports the concept that IDH1(R132MUT) gliomas arise from a neural precursor population that is spatially and temporally restricted in the brain. The observed patterns of DNA sequence, methylation, and copy number alterations support a model of ordered molecular evolution of IDH1(R132MUT) GBM in which the appearance of mutant IDH1 protein is an initial event, followed by production of p53 mutant protein, and finally by copy number alterations of PTEN and EGFR. CONCLUSION Although histologically similar, GBMs arising with and without IDH1(R132MUT) appear to represent distinct disease entities that arise from separate cell types of origin as the result of largely nonoverlapping sets of molecular events. Optimal clinical management should account for the distinction between these GBM disease subtypes.


American Journal of Neuroradiology | 2012

Relationship between tumor enhancement, edema, IDH1 mutational status, MGMT promoter methylation, and survival in glioblastoma.

J.A. Carrillo; Albert Lai; Phioanh L. Nghiemphu; Hyun J. Kim; Heidi S. Phillips; Samir Kharbanda; Parham Moftakhar; S. Lalaezari; William H. Yong; Benjamin M. Ellingson; T. Cloughesy; W.B. Pope

IDH1 mutations and MGMT promoter methylation correlate with better prognosis and longer survival in patients with glioblastomas. This article looks at the relationship between edema and contrast enhancement and these features. The authors studied 202 glioblastomas and found that patients with methylated tumors with no or little edema had longer survival rates. All IDH1 mutant tumors were located in the frontal lobes and were mostly nonenhancing. It seems that common anatomic features may provide insight into the molecular behavior of some glioblastomas. BACKGROUND AND PURPOSE: Both IDH1 mutation and MGMT promoter methylation are associated with longer survival. We investigated the ability of imaging correlates to serve as noninvasive biomarkers for these molecularly defined GBM subtypes. MATERIALS AND METHODS: MR imaging from 202 patients with GBM was retrospectively assessed for nonenhancing tumor and edema among other imaging features. IDH1 mutational and MGMT promoter methylation status were determined by DNA sequencing and methylation-specific PCR, respectively. Overall survival was determined by using a multivariate Cox model and the Kaplan-Meier method with a log rank test. A logistic regression model followed by ROC analysis was used to classify the IDH1 mutation and methylation status by using imaging features. RESULTS: MGMT promoter methylation and IDH1 mutation were associated with longer median survival. Edema levels stratified survival for methylated but not unmethylated tumors. Median survival for methylated tumors with little/no edema was 2476 days (95% CI, 795), compared with 586 days (95% CI, 507–654) for unmethylated tumors or tumors with edema. All IDH1 mutant tumors were nCET positive, and most (11/14, 79%) were located in the frontal lobe. Imaging features including larger tumor size and nCET could be used to determine IDH1 mutational status with 97.5% accuracy, but poorly predicted MGMT promoter methylation. CONCLUSIONS: Imaging features are potentially predictive of IDH1 mutational status but were poorly correlated with MGMT promoter methylation. Edema stratifies survival in MGMT promoter methylated but not in unmethylated tumors; patients with methylated tumors with little or no edema have particularly long survival.


Journal of Clinical Oncology | 2015

Patients With Proneural Glioblastoma May Derive Overall Survival Benefit From the Addition of Bevacizumab to First-Line Radiotherapy and Temozolomide: Retrospective Analysis of the AVAglio Trial

Thomas Sandmann; Richard Bourgon; Josep Garcia; Congfen Li; Timothy F. Cloughesy; Olivier Chinot; Wolfgang Wick; Ryo Nishikawa; Warren P. Mason; Roger Henriksson; Frank Saran; Albert Lai; Nicola Moore; Samir Kharbanda; Franklin Peale; Priti Hegde; Lauren E. Abrey; Heidi S. Phillips; Carlos Bais

PURPOSE The AVAglio (Avastin in Glioblastoma) and RTOG-0825 randomized, placebo-controlled phase III trials in newly diagnosed glioblastoma reported prolonged progression-free survival (PFS), but not overall survival (OS), with the addition of bevacizumab to radiotherapy plus temozolomide. To establish whether certain patient subgroups derived an OS benefit from the addition of bevacizumab to first-line standard-of-care therapy, AVAglio patients were retrospectively evaluated for molecular subtype, and bevacizumab efficacy was assessed for each patient subgroup. PATIENTS AND METHODS A total of 349 pretreatment specimens (bevacizumab arm, n = 171; placebo arm, n = 178) from AVAglio patients (total, N = 921) were available for biomarker analysis. Samples were profiled for gene expression and isocitrate dehydrogenase 1 (IDH1) mutation status and classified into previously identified molecular subtypes. PFS and OS were assessed within each subtype. RESULTS A multivariable analysis accounting for prognostic covariates revealed that bevacizumab conferred a significant OS advantage versus placebo for patients with proneural IDH1 wild-type tumors (17.1 v 12.8 months, respectively; hazard ratio, 0.43; 95% CI, 0.26 to 0.73; P = .002). This analysis also revealed an interaction between the proneural subtype biomarker and treatment arm (P = .023). The group of patients with mesenchymal and proneural tumors derived a PFS benefit from bevacizumab compared with placebo; however, this translated to an OS benefit in the proneural subset only. CONCLUSION Retrospective analysis of AVAglio data suggests that patients with IDH1 wild-type proneural glioblastoma may derive an OS benefit from first-line bevacizumab treatment. The predictive value of the proneural subtype observed in AVAglio should be validated in an independent data set.


Proceedings of the National Academy of Sciences of the United States of America | 2007

Identification of IGF2 signaling through phosphoinositide-3-kinase regulatory subunit 3 as a growth-promoting axis in glioblastoma.

Liliana Soroceanu; Samir Kharbanda; Ruihuan Chen; Robert Soriano; Kenneth D. Aldape; Anjan Misra; Jiping Zha; William F. Forrest; Janice Nigro; Zora Modrusan; Burt G. Feuerstein; Heidi S. Phillips

Amplification or overexpression of growth factor receptors is a frequent occurrence in malignant gliomas. Using both expression profiling and in situ hybridization, we identified insulin-like growth factor 2 (IGF2) as a marker for a subset of glioblastomas (GBMs) that lack amplification or overexpression of EGF receptor. Among 165 primary high-grade astrocytomas, 13% of grade IV tumors and 2% of grade III tumors expressed IGF2 mRNA levels >50-fold the sample population median. IGF2-overexpressing tumors frequently displayed PTEN loss, were highly proliferative, exhibited strong staining for phospho-Akt, and belonged to a subclass of GBMs characterized by poor survival. Using a serum-free culture system, we discovered that IGF2 can substitute for EGF to support the growth of GBM-derived neurospheres. The growth-promoting effects of IGF2 were mediated by the insulin-like growth factor receptor 1 and phosphoinositide-3-kinase regulatory subunit 3 (PIK3R3), a regulatory subunit of phosphoinositide 3-kinase that shows genomic gains in some highly proliferative GBM cases. PIK3R3 knockdown inhibited IGF2-induced growth of GBM-derived neurospheres. The current results provide evidence that the IGF2–PIK3R3 signaling axis is involved in promoting the growth of a subclass of highly aggressive human GBMs that lack EGF receptor amplification. Our data underscore the importance of the phosphoinositide 3-kinase/Akt pathway for growth of high-grade gliomas and suggest that multiple molecular alterations that activate this signaling cascade may promote tumorigenesis. Further, these findings highlight the parallels between growth factors or receptors that are overexpressed in GBMs and those that support in vitro growth of tumor-derived stem-like cells.


Glia | 2011

A distinct subset of glioma cell lines with stem cell-like properties reflects the transcriptional phenotype of glioblastomas and overexpresses CXCR4 as therapeutic target

Alexander Schulte; Hauke S. Günther; Heidi S. Phillips; Dirk Kemming; Tobias Martens; Samir Kharbanda; Robert Soriano; Zora Modrusan; Svenja Zapf; Manfred Westphal; Katrin Lamszus

Glioblastomas contain stem‐like cells that can be maintained in vitro using specific serum‐free conditions. We investigated whether glioblastoma stem‐like (GS) cell lines preserve the expression phenotype of human glioblastomas more closely than conventional glioma cell lines. Expression profiling revealed that a distinct subset of GS lines, which displayed a full stem‐like phenotype (GSf), mirrored the expression signature of glioblastomas more closely than either other GS lines or cell lines grown in serum. GSf lines are highly tumorigenic and invasive in vivo, express CD133, grow spherically in vitro, are multipotent and display a Proneural gene expression signature, thus recapitulating key functional and transcriptional aspects of human glioblastomas. In contrast, GS lines with a restricted stem‐like phenotype exhibited expression signatures more similar to conventional cell lines than to original patient tumors, suggesting that the transcriptional resemblance between GS lines and tumors is associated with different degrees of “stemness”. Among markers overexpressed in patient tumors and GSf lines, we identified CXCR4 as a potential therapeutic target. GSf lines contained a minor population of CXCR4hi cells, a subfraction of which coexpressed CD133 and was expandable by hypoxia, whereas conventional cell lines contained only CXCR4lo cells. Convection‐enhanced local treatment with AMD3100, a specific CXCR4 antagonist, inhibited the highly invasive growth of GS xenografts in vivo and cell migration in vitro. We thus demonstrate the utility of GSf lines in testing therapeutic agents and validate CXCR4 as a target to block the growth of invasive tumor‐initiating glioma stem cells in vivo.


Proceedings of the National Academy of Sciences of the United States of America | 2007

Epigenome scans and cancer genome sequencing converge on WNK2, a kinase-independent suppressor of cell growth

Chibo Hong; K. Scott Moorefield; Peter Jun; Kenneth D. Aldape; Samir Kharbanda; Heidi S. Phillips; Joseph F. Costello

Human cancer genome and epigenome projects aim to identify new cancer genes and targets for therapy that have been overlooked by conventional approaches. Here we integrated large-scale genomics and epigenomics of 31 human infiltrative gliomas and identified low-frequency deletion and highly recurrent epigenetic silencing of WNK2, encoding a putative serine/threonine kinase. Prior cancer genome sequencing projects also identified point mutations in WNK1–4, suggesting that WNK family genes may have a role in cancers. We observed consistent gene silencing in tumors with dense aberrant methylation across 1.3 kb of the CpG island but more variable expression when the 5′-most region remained unmethylated. This primary tumor data fit well with WNK2 promoter analysis, which showed strong promoter activity in the 5′-most region, equivalent to the simian virus 40 promoter, but no activity in the 3′ region. WT WNK2 exhibited autophosphorylation and protein kinase activity that was enhanced in cells exposed to hypertonic conditions, similar to WNK1. WNK2 inhibited up to 78% of colony formation by glioma cells but in an unexpectedly kinase-independent manner. The WNK2 silencing by epigenetic mechanisms was significantly associated (P < 0.01) with a known genetic signature of chemosensitive oligodendroglial tumors, 1p and 19q deletion, in two small but independent tumor sets. Taken together, the epigenetic silencing, occasional deletion and point mutation, and functional assessment suggest that aberrations of WNK2 may contribute to unregulated tumor cell growth. Thus, our integrated genetic and epigenetic approach might be useful to identify genes that are widely relevant to cancer, even when genetic alterations of the locus are infrequent.


Clinical Cancer Research | 2012

Targeting the PI3K Pathway in the Brain - Efficacy of a PI3K Inhibitor Optimized to Cross the Blood-Brain Barrier

Laurent Salphati; Timothy P. Heffron; Bruno Alicke; Merry Nishimura; Kai H. Barck; Richard A. D. Carano; Jonathan Cheong; Kyle A. Edgar; Joan M. Greve; Samir Kharbanda; Hartmut Koeppen; Shari Lau; Leslie Lee; Jodie Pang; Emile Plise; Jenny L. Pokorny; Hani Bou Reslan; Jann N. Sarkaria; Jeffrey Wallin; Xiaolin Zhang; Stephen E. Gould; Alan G. Olivero; Heidi S. Phillips

Purpose: Glioblastoma (GBM), the most common primary brain tumor in adults, presents a high frequency of alteration in the PI3K pathway. Our objectives were to identify a dual PI3K/mTOR inhibitor optimized to cross the blood–brain barrier (BBB) and characterize its brain penetration, pathway modulation in the brain and efficacy in orthotopic xenograft models of GBM. Experimental Design: Physicochemical properties of PI3K inhibitors were optimized using in silico tools, leading to the identification of GNE-317. This compound was tested in cells overexpressing P-glycoprotein (P-gp) or breast cancer resistance protein (BCRP). Following administration to mice, GNE-317 plasma and brain concentrations were determined, and phosphorylated biomarkers (pAkt, p4EBP1, and pS6) were measured to assess PI3K pathway suppression in the brain. GNE-317 efficacy was evaluated in the U87, GS2, and GBM10 orthotopic models of GBM. Results: GNE-317 was identified as having physicochemical properties predictive of low efflux by P-gp and BCRP. Studies in transfected MDCK cells showed that GNE-317 was not a substrate of either transporter. GNE-317 markedly inhibited the PI3K pathway in mouse brain, causing 40% to 90% suppression of the pAkt and pS6 signals up to 6-hour postdose. GNE-317 was efficacious in the U87, GS2, and GBM10 orthotopic models, achieving tumor growth inhibition of 90% and 50%, and survival benefit, respectively. Conclusions: These results indicated that specific optimization of PI3K inhibitors to cross the BBB led to potent suppression of the PI3K pathway in healthy brain. The efficacy of GNE-317 in 3 intracranial models of GBM suggested that this compound could be effective in the treatment of GBM. Clin Cancer Res; 18(22); 6239–48. ©2012 AACR.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Hominoid-specific enzyme GLUD2 promotes growth of IDH1R132H glioma

Ruihuan Chen; Merry Nishimura; Samir Kharbanda; Frank Peale; Yuzhong Deng; Anneleen Daemen; William F. Forrest; Mandy Kwong; Maj Hedehus; Georgia Hatzivassiliou; Lori Friedman; Heidi S. Phillips

Significance Mutation of isocitrate dehydrogenase 1 (IDH1) is believed to be the initiating event for the majority of secondary glioblastomas and lower-grade diffuse gliomas; however, the basis for tissue specificity of oncogenesis initiated by IDH1 mutation has not been apparent. We report evidence to suggest that specialization of human neocortex for glutaminergic neurotransmission provides a metabolic niche particularly suited for growth of IDH1R132H glioma. Our findings reveal that IDH1-mutant enzyme challenges growth of murine glioma progenitor cells but that these cells thrive if they are engineered to express the hominoid-specific brain enzyme GLUD2, a mitochondrial enzyme that converts glutamate to alpha-ketoglutarate in human cortex. The current findings raise the possibility that evolutionary changes contributing to human cognitive abilities may have conferred vulnerability to brain tumors driven by IDH1 mutation. Somatic mutation of isocitrate dehydrogenase 1 (IDH1) is now recognized as the most common initiating event for secondary glioblastoma, a brain tumor type arising with high frequency in the frontal lobe. A puzzling feature of IDH1 mutation is the selective manifestation of glioma as the only neoplasm frequently associated with early postzygotic occurrence of this genomic alteration. We report here that IDH1R132H exhibits a growth-inhibitory effect that is abrogated in the presence of glutamate dehydrogenase 2 (GLUD2), a hominoid-specific enzyme purportedly optimized to facilitate glutamate turnover in human forebrain. Using murine glioma progenitor cells, we demonstrate that IDH1R132H exerts a growth-inhibitory effect that is paralleled by deficiency in metabolic flux from glucose and glutamine to lipids. Examining human gliomas, we find that glutamate dehydrogenase 1 (GLUD1) and GLUD2 are overexpressed in IDH1-mutant tumors and that orthotopic growth of an IDH1-mutant glioma line is inhibited by knockdown of GLUD1/2. Strikingly, introduction of GLUD2 into murine glioma progenitor cells reverses deleterious effects of IDH1 mutation on metabolic flux and tumor growth. Further, we report that glutamate, a substrate of GLUD2 and a neurotransmitter abundant in mammalian neocortex, can support growth of glioma progenitor cells irrespective of IDH1 mutation status. These findings suggest that specialization of human neocortex for high glutamate neurotransmitter flux creates a metabolic niche conducive to growth of IDH1 mutant tumors.


American Journal of Neuroradiology | 2012

Differential Gene Expression in Glioblastoma Defined by ADC Histogram Analysis: Relationship to Extracellular Matrix Molecules and Survival

Whitney B. Pope; Leili Mirsadraei; Albert Lai; A. Eskin; J. Qiao; Hyun J. Kim; Benjamin M. Ellingson; Phioanh L. Nghiemphu; Samir Kharbanda; R.H. Soriano; S.F. Nelson; William H. Yong; Heidi S. Phillips; Timothy F. Cloughesy

BACKGROUND AND PURPOSE: ADC histogram analysis can stratify outcomes in patients with GBM treated with bevacizumab. Therefore, we compared gene expression between high-versus-low ADC tumors to identify gene expression modules that could underlie this difference and impact patient prognosis. MATERIALS AND METHODS: Up-front bevacizumab-treated patients (N = 38) with newly diagnosed glioblastoma were analyzed by using an ADC histogram approach based on enhancing tumor. Using microarrays, we compared gene expression in high-versus-low ADC tumors in patients subsequently treated with bevacizumab. Tissue sections from a subset of tumors were stained for collagen and collagen-binding proteins. Progression-free and overall survival was determined by using Cox proportional hazard ratios and the Kaplan-Meier method with the log rank test. RESULTS: A total of 13 genes were expressed at 2-fold or greater levels in high- compared with low-ADC tumors at the P < .05 level. Of these, 6 encode for collagen or collagen-binding proteins. High gene expression for the collagen-binding protein decorin was associated with shorter survival (HR, 2.5; P = .03). The pattern and degree of collagen staining were highly variable in both high- and low-ADC tumors. CONCLUSIONS: High-ADC GBMs show greater levels of ECM protein gene expression compared with low-ADC GBMs. It is unclear whether this translates to the accumulation of higher levels of the encoded proteins. However, because ECM molecules could contribute to a proinvasive phenotype, this relationship merits further investigation.

Collaboration


Dive into the Samir Kharbanda's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Albert Lai

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kenneth D. Aldape

Princess Margaret Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Anjan Misra

Barrow Neurological Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Burt G. Feuerstein

Barrow Neurological Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge