Network


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

Hotspot


Dive into the research topics where Igor Vivanco is active.

Publication


Featured researches published by Igor Vivanco.


Nature Reviews Cancer | 2002

The phosphatidylinositol 3-Kinase AKT pathway in human cancer.

Igor Vivanco; Charles L. Sawyers

One signal that is overactivated in a wide range of tumour types is the production of a phospholipid, phosphatidylinositol (3,4,5) trisphosphate, by phosphatidylinositol 3-kinase (PI3K). This lipid and the protein kinase that is activated by it — AKT — trigger a cascade of responses, from cell growth and proliferation to survival and motility, that drive tumour progression. Small-molecule therapeutics that block PI3K signalling might deal a severe blow to cancer cells by blocking many aspects of the tumour-cell phenotype.


Nature | 2002

The phosphatidylinositol 3-Kinase|[ndash]|AKT pathway in human cancer

Igor Vivanco; Charles L. Sawyers

One signal that is overactivated in a wide range of tumour types is the production of a phospholipid, phosphatidylinositol (3,4,5) trisphosphate, by phosphatidylinositol 3-kinase (PI3K). This lipid and the protein kinase that is activated by it — AKT — trigger a cascade of responses, from cell growth and proliferation to survival and motility, that drive tumour progression. Small-molecule therapeutics that block PI3K signalling might deal a severe blow to cancer cells by blocking many aspects of the tumour-cell phenotype.


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

Assessing the significance of chromosomal aberrations in cancer: Methodology and application to glioma

Rameen Beroukhim; Gad Getz; Leia Nghiemphu; Jordi Barretina; Teli Hsueh; David Linhart; Igor Vivanco; Jeffrey C. Lee; Julie H. Huang; Sethu Alexander; Jinyan Du; Tweeny R. Kau; Roman K. Thomas; Kinjal Shah; Horacio Soto; Sven Perner; John R. Prensner; Ralph DeBiasi; Francesca Demichelis; Charlie Hatton; Mark A. Rubin; Levi A. Garraway; Stan F. Nelson; Linda M. Liau; Paul S. Mischel; T. Cloughesy; Matthew Meyerson; Todd Golub; Eric S. Lander; Ingo K. Mellinghoff

Comprehensive knowledge of the genomic alterations that underlie cancer is a critical foundation for diagnostics, prognostics, and targeted therapeutics. Systematic efforts to analyze cancer genomes are underway, but the analysis is hampered by the lack of a statistical framework to distinguish meaningful events from random background aberrations. Here we describe a systematic method, called Genomic Identification of Significant Targets in Cancer (GISTIC), designed for analyzing chromosomal aberrations in cancer. We use it to study chromosomal aberrations in 141 gliomas and compare the results with two prior studies. Traditional methods highlight hundreds of altered regions with little concordance between studies. The new approach reveals a highly concordant picture involving ≈35 significant events, including 16–18 broad events near chromosome-arm size and 16–21 focal events. Approximately half of these events correspond to known cancer-related genes, only some of which have been previously tied to glioma. We also show that superimposed broad and focal events may have different biological consequences. Specifically, gliomas with broad amplification of chromosome 7 have properties different from those with overlapping focalEGFR amplification: the broad events act in part through effects on MET and its ligand HGF and correlate with MET dependence in vitro. Our results support the feasibility and utility of systematic characterization of the cancer genome.


Journal of Clinical Investigation | 2009

Dual targeting of EGFR can overcome a major drug resistance mutation in mouse models of EGFR mutant lung cancer

Lucia Regales; Yixuan Gong; Ronglai Shen; Elisa de Stanchina; Igor Vivanco; Aviva Goel; Jason A. Koutcher; Maria Spassova; Ouathek Ouerfelli; Ingo K. Mellinghoff; Maureen F. Zakowski; Katerina Politi; William Pao

EGFR is a major anticancer drug target in human epithelial tumors. One effective class of agents is the tyrosine kinase inhibitors (TKIs), such as gefitinib and erlotinib. These drugs induce dramatic responses in individuals with lung adenocarcinomas characterized by mutations in exons encoding the EGFR tyrosine kinase domain, but disease progression invariably occurs. A major reason for such acquired resistance is the outgrowth of tumor cells with additional TKI-resistant EGFR mutations. Here we used relevant transgenic mouse lung tumor models to evaluate strategies to overcome the most common EGFR TKI resistance mutation, T790M. We treated mice bearing tumors harboring EGFR mutations with a variety of anticancer agents, including a new irreversible EGFR TKI that is under development (BIBW-2992) and the EGFR-specific antibody cetuximab. Surprisingly, we found that only the combination of both agents together induced dramatic shrinkage of erlotinib-resistant tumors harboring the T790M mutation, because together they efficiently depleted both phosphorylated and total EGFR. We suggest that these studies have immediate therapeutic implications for lung cancer patients, as dual targeting with cetuximab and a second-generation EGFR TKI may be an effective strategy to overcome T790M-mediated drug resistance. Moreover, this approach could serve as an important model for targeting other receptor tyrosine kinases activated in human cancers.


PLOS Medicine | 2006

Epidermal Growth Factor Receptor Activation in Glioblastoma through Novel Missense Mutations in the Extracellular Domain

Jeffrey C. Lee; Igor Vivanco; Rameen Beroukhim; Julie H.Y. Huang; Whei Feng; Ralph DeBiasi; Koji Yoshimoto; J. King; Phioanh L. Nghiemphu; Yuki Yuza; Qing-Qing Xu; Heidi Greulich; Roman K. Thomas; J. Guillermo Paez; Timothy C. Peck; David Linhart; Karen A. Glatt; Gad Getz; Robert C. Onofrio; Liuda Ziaugra; Ross L. Levine; Stacey Gabriel; Tomohiro Kawaguchi; Keith O'Neill; Haumith Khan; Linda M. Liau; Stanley F. Nelson; P. Nagesh Rao; Paul S. Mischel; Russell O. Pieper

Background Protein tyrosine kinases are important regulators of cellular homeostasis with tightly controlled catalytic activity. Mutations in kinase-encoding genes can relieve the autoinhibitory constraints on kinase activity, can promote malignant transformation, and appear to be a major determinant of response to kinase inhibitor therapy. Missense mutations in the EGFR kinase domain, for example, have recently been identified in patients who showed clinical responses to EGFR kinase inhibitor therapy. Methods and Findings Encouraged by the promising clinical activity of epidermal growth factor receptor (EGFR) kinase inhibitors in treating glioblastoma in humans, we have sequenced the complete EGFR coding sequence in glioma tumor samples and cell lines. We identified novel missense mutations in the extracellular domain of EGFR in 13.6% (18/132) of glioblastomas and 12.5% (1/8) of glioblastoma cell lines. These EGFR mutations were associated with increased EGFR gene dosage and conferred anchorage-independent growth and tumorigenicity to NIH-3T3 cells. Cells transformed by expression of these EGFR mutants were sensitive to small-molecule EGFR kinase inhibitors. Conclusions Our results suggest extracellular missense mutations as a novel mechanism for oncogenic EGFR activation and may help identify patients who can benefit from EGFR kinase inhibitors for treatment of glioblastoma.


Nature Genetics | 2010

Somatic mutations of the Parkinson's disease–associated gene PARK2 in glioblastoma and other human malignancies

Selvaraju Veeriah; Barry S. Taylor; Shasha Meng; Fang Fang; Emrullah Yilmaz; Igor Vivanco; Manickam Janakiraman; Nikolaus Schultz; Aphrothiti J. Hanrahan; William Pao; Marc Ladanyi; Chris Sander; Adriana Heguy; Eric C. Holland; Philip B. Paty; Paul S. Mischel; Linda M. Liau; Timothy F. Cloughesy; Ingo K. Mellinghoff; David B. Solit; Timothy A. Chan

Mutation of the gene PARK2, which encodes an E3 ubiquitin ligase, is the most common cause of early-onset Parkinsons disease. In a search for multisite tumor suppressors, we identified PARK2 as a frequently targeted gene on chromosome 6q25.2–q27 in cancer. Here we describe inactivating somatic mutations and frequent intragenic deletions of PARK2 in human malignancies. The PARK2 mutations in cancer occur in the same domains, and sometimes at the same residues, as the germline mutations causing familial Parkinsons disease. Cancer-specific mutations abrogate the growth-suppressive effects of the PARK2 protein. PARK2 mutations in cancer decrease PARK2s E3 ligase activity, compromising its ability to ubiquitinate cyclin E and resulting in mitotic instability. These data strongly point to PARK2 as a tumor suppressor on 6q25.2–q27. Thus, PARK2, a gene that causes neuronal dysfunction when mutated in the germline, may instead contribute to oncogenesis when altered in non-neuronal somatic cells.


Cancer Research | 2006

Mammalian target of rapamycin inhibition promotes response to epidermal growth factor receptor kinase inhibitors in PTEN-deficient and PTEN-intact glioblastoma cells

Maria Y. Wang; Kan V. Lu; Shaojun Zhu; Ederlyn Q. Dia; Igor Vivanco; Gregory M. Shackleford; Webster K. Cavenee; Ingo K. Mellinghoff; Timothy F. Cloughesy; Charles L. Sawyers; Paul S. Mischel

The epidermal growth factor receptor (EGFR) is commonly amplified, overexpressed, and mutated in glioblastoma, making it a compelling molecular target for therapy. We have recently shown that coexpression of EGFRvIII and PTEN protein by glioblastoma cells is strongly associated with clinical response to EGFR kinase inhibitor therapy. PTEN loss, by dissociating inhibition of the EGFR from downstream phosphatidylinositol 3-kinase (PI3K) pathway inhibition, seems to act as a resistance factor. Because 40% to 50% of glioblastomas are PTEN deficient, a critical challenge is to identify strategies that promote responsiveness to EGFR kinase inhibitors in patients whose tumors lack PTEN. Here, we show that the mammalian target of rapamycin (mTOR) inhibitor rapamycin enhances the sensitivity of PTEN-deficient tumor cells to the EGFR kinase inhibitor erlotinib. In two isogenic model systems (U87MG glioblastoma cells expressing EGFR, EGFRvIII, and PTEN in relevant combinations, and SF295 glioblastoma cells in which PTEN protein expression has been stably restored), we show that combined EGFR/mTOR kinase inhibition inhibits tumor cell growth and has an additive effect on inhibiting downstream PI3K pathway signaling. We also show that combination therapy provides added benefit in promoting cell death in PTEN-deficient tumor cells. These studies provide strong rationale for combined mTOR/EGFR kinase inhibitor therapy in glioblastoma patients, particularly those with PTEN-deficient tumors.


Cancer Discovery | 2012

Differential sensitivity of glioma- versus lung cancer-specific EGFR mutations to EGFR kinase inhibitors.

Igor Vivanco; H. Ian Robins; Daniel Rohle; Carl Campos; Christian Grommes; Phioanh L. Nghiemphu; Sara Kubek; Barbara Oldrini; Milan G. Chheda; Nicolas Yannuzzi; Hui Tao; Shaojun Zhu; Akio Iwanami; Daisuke Kuga; Julie Dang; Alicia Pedraza; Cameron Brennan; Adriana Heguy; Linda M. Liau; Frank S. Lieberman; W. K. Alfred Yung; Mark R. Gilbert; David A. Reardon; Jan Drappatz; Patrick Y. Wen; Kathleen R. Lamborn; Susan M. Chang; Michael D. Prados; Howard A. Fine; Steve Horvath

UNLABELLED Activation of the epidermal growth factor receptor (EGFR) in glioblastoma (GBM) occurs through mutations or deletions in the extracellular (EC) domain. Unlike lung cancers with EGFR kinase domain (KD) mutations, GBMs respond poorly to the EGFR inhibitor erlotinib. Using RNAi, we show that GBM cells carrying EGFR EC mutations display EGFR addiction. In contrast to KD mutants found in lung cancer, glioma-specific EGFR EC mutants are poorly inhibited by EGFR inhibitors that target the active kinase conformation (e.g., erlotinib). Inhibitors that bind to the inactive EGFR conformation, however, potently inhibit EGFR EC mutants and induce cell death in EGFR-mutant GBM cells. Our results provide first evidence for single kinase addiction in GBM and suggest that the disappointing clinical activity of first-generation EGFR inhibitors in GBM versus lung cancer may be attributed to the different conformational requirements of mutant EGFR in these 2 cancer types. SIGNIFICANCE Approximately 40% of human glioblastomas harbor oncogenic EGFR alterations, but attempts to therapeutically target EGFR with first-generation EGFR kinase inhibitors have failed. Here, we demonstrate selective sensitivity of glioma-specific EGFR mutants to ATP-site competitive EGFR kinase inhibitors that target the inactive conformation of the catalytic domain.


Proceedings of the National Academy of Sciences of the United States of America | 2009

The tyrosine phosphatase PTPRD is a tumor suppressor that is frequently inactivated and mutated in glioblastoma and other human cancers

Selvaraju Veeriah; Cameron Brennan; Shasha Meng; Bhuvanesh Singh; James A. Fagin; David B. Solit; Philip B. Paty; D. Rohle; Igor Vivanco; Juliann Chmielecki; William Pao; Marc Ladanyi; William L. Gerald; Linda M. Liau; Timothy Cloughesy; Paul S. Mischel; Chris Sander; Barry S. Taylor; Nikolaus Schultz; John Major; Adriana Heguy; Fang Fang; Ingo K. Mellinghoff; Timothy A. Chan

Tyrosine phosphorylation plays a critical role in regulating cellular function and is a central feature in signaling cascades involved in oncogenesis. The regulation of tyrosine phosphorylation is coordinately controlled by kinases and phosphatases (PTPs). Whereas activation of tyrosine kinases has been shown to play vital roles in tumor development, the role of PTPs is much less well defined. Here, we show that the receptor protein tyrosine phosphatase delta (PTPRD) is frequently inactivated in glioblastoma multiforme (GBM), a deadly primary neoplasm of the brain. PTPRD is a target of deletion in GBM, often via focal intragenic loss. In GBM tumors that do not possess deletions in PTPRD, the gene is frequently subject to cancer-specific epigenetic silencing via promoter CpG island hypermethylation (37%). Sequencing of the PTPRD gene in GBM and other primary human tumors revealed that the gene is mutated in 6% of GBMs, 13% of head and neck squamous cell carcinomas, and in 9% of lung cancers. These mutations were deleterious. In total, PTPRD inactivation occurs in >50% of GBM tumors, and loss of expression predicts for poor prognosis in glioma patients. Wild-type PTPRD inhibits the growth of GBM and other tumor cells, an effect not observed with PTPRD alleles harboring cancer-specific mutations. Human astrocytes lacking PTPRD exhibited increased growth. PTPRD was found to dephosphorylate the oncoprotein STAT3. These results implicate PTPRD as a tumor suppressor on chromosome 9p that is involved in the development of GBMs and multiple human cancers.


Cancer Research | 2007

Murine Cell Lines Derived from Pten Null Prostate Cancer Show the Critical Role of PTEN in Hormone Refractory Prostate Cancer Development

Jing Jiao; Shunyou Wang; Rong Qiao; Igor Vivanco; Philip A. Watson; Charles L. Sawyers; Hong Wu

PTEN mutations are among the most frequent genetic alterations found in human prostate cancers. Our previous works suggest that although precancerous lesions were found in Pten heterozygous mice, cancer progression and metastasis only happened when both alleles of Pten were deleted. To understand the molecular mechanisms underlying the role of PTEN in prostate cancer control, we generated two pairs of isogenic, androgen receptor (AR)-positive prostate epithelial lines from intact conditional Pten knock-out mice that are either heterozygous (PTEN-P2 and -P8) or homozygous (PTEN-CaP2 and PTEN-CaP8) for Pten deletion. Further characterization of these cells showed that loss of the second allele of Pten leads to increased anchorage-independent growth in vitro and tumorigenesis in vivo without obvious structural or numerical chromosome changes based on SKY karyotyping analysis. Despite no prior exposure to hormone ablation therapy, Pten null cells are tumorigenic in both male and female severe combined immunodeficiency mice. Furthermore, knocking down PTEN can convert the androgen-dependent Myc-CaP cell into androgen independence, suggesting that PTEN intrinsically controls androgen responsiveness, a critical step in the development of hormone refractory prostate cancer. Importantly, knocking down AR by shRNA in Pten null cells reverses androgen-independent growth in vitro and partially inhibited tumorigenesis in vivo, indicating that PTEN-controlled prostate tumorigenesis is AR dependent. These cell lines will serve as useful tools for understanding signaling pathways controlled by PTEN and elucidating the molecular mechanisms involved in hormone refractory prostate cancer formation.

Collaboration


Dive into the Igor Vivanco's collaboration.

Top Co-Authors

Avatar

Ingo K. Mellinghoff

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Paul S. Mischel

Ludwig Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Charles L. Sawyers

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Linda M. Liau

University of California

View shared research outputs
Top Co-Authors

Avatar

Barbara Oldrini

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Barry S. Taylor

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Carl Campos

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge