Network


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

Hotspot


Dive into the research topics where Carl Campos is active.

Publication


Featured researches published by Carl Campos.


Nature | 2012

IDH1 mutation is sufficient to establish the glioma hypermethylator phenotype

Sevin Turcan; Daniel Rohle; Anuj Goenka; Logan A. Walsh; Fang Fang; Emrullah Yilmaz; Carl Campos; Armida W. M. Fabius; Chao Lu; Patrick S. Ward; Craig B. Thompson; Andrew Kaufman; Olga A. Guryanova; Ross L. Levine; Adriana Heguy; Agnes Viale; Luc G. T. Morris; Jason T. Huse; Ingo K. Mellinghoff; Timothy A. Chan

Both genome-wide genetic and epigenetic alterations are fundamentally important for the development of cancers, but the interdependence of these aberrations is poorly understood. Glioblastomas and other cancers with the CpG island methylator phenotype (CIMP) constitute a subset of tumours with extensive epigenomic aberrations and a distinct biology. Glioma CIMP (G-CIMP) is a powerful determinant of tumour pathogenicity, but the molecular basis of G-CIMP remains unresolved. Here we show that mutation of a single gene, isocitrate dehydrogenase 1 (IDH1), establishes G-CIMP by remodelling the methylome. This remodelling results in reorganization of the methylome and transcriptome. Examination of the epigenome of a large set of intermediate-grade gliomas demonstrates a distinct G-CIMP phenotype that is highly dependent on the presence of IDH mutation. Introduction of mutant IDH1 into primary human astrocytes alters specific histone marks, induces extensive DNA hypermethylation, and reshapes the methylome in a fashion that mirrors the changes observed in G-CIMP-positive lower-grade gliomas. Furthermore, the epigenomic alterations resulting from mutant IDH1 activate key gene expression programs, characterize G-CIMP-positive proneural glioblastomas but not other glioblastomas, and are predictive of improved survival. Our findings demonstrate that IDH mutation is the molecular basis of CIMP in gliomas, provide a framework for understanding oncogenesis in these gliomas, and highlight the interplay between genomic and epigenomic changes in human cancers.


Science | 2013

An Inhibitor of Mutant IDH1 Delays Growth and Promotes Differentiation of Glioma Cells

D. Rohle; Janeta Popovici-Muller; Nicolaos Palaskas; Sevin Turcan; Christian Grommes; Carl Campos; Jennifer Tsoi; Owen Clark; Barbara Oldrini; Evangelia Komisopoulou; Kaiko Kunii; Alicia Pedraza; Stefanie Schalm; Lee Silverman; Alexandra Miller; Fang Wang; Hua Yang; Yue Chen; Andrew Kernytsky; Marc K. Rosenblum; Wei Liu; Scott A. Biller; Shinsan M. Su; Cameron Brennan; Timothy A. Chan; Thomas G. Graeber; Katharine E. Yen; Ingo K. Mellinghoff

IDHology Among the most exciting drug targets to emerge from cancer genome sequencing projects are two related metabolic enzymes, isocitrate dehydrogenases 1 and 2 (IDH1, IDH2). Mutations in the IDH1 and IDH2 genes are common in certain types of human cancer. Whether inhibition of mutant IDH activity might offer therapeutic benefits is unclear (see the Perspective by Kim and DeBerardinis). F. Wang et al. (p. 622, published online 4 April) isolated a small molecule that selectively inhibits mutant IDH2, describe the structural details of its binding to the mutant enzyme, and show that this compound suppresses the growth of patient-derived leukemia cells harboring the IDH2 mutation. Rohle et al. (p. 626, published online 4 April) show that a small molecule inhibitor of IDH1 selectively slows the growth of patient-derived brain tumor cells with the IDH1 mutation. A small molecule that inhibits a mutant enzyme in tumors slows malignant growth by inducing cancer cell differentiation. [Also see Perspective by Kim and DeBerardinis] The recent discovery of mutations in metabolic enzymes has rekindled interest in harnessing the altered metabolism of cancer cells for cancer therapy. One potential drug target is isocitrate dehydrogenase 1 (IDH1), which is mutated in multiple human cancers. Here, we examine the role of mutant IDH1 in fully transformed cells with endogenous IDH1 mutations. A selective R132H-IDH1 inhibitor (AGI-5198) identified through a high-throughput screen blocked, in a dose-dependent manner, the ability of the mutant enzyme (mIDH1) to produce R-2-hydroxyglutarate (R-2HG). Under conditions of near-complete R-2HG inhibition, the mIDH1 inhibitor induced demethylation of histone H3K9me3 and expression of genes associated with gliogenic differentiation. Blockade of mIDH1 impaired the growth of IDH1-mutant—but not IDH1–wild-type—glioma cells without appreciable changes in genome-wide DNA methylation. These data suggest that mIDH1 may promote glioma growth through mechanisms beyond its well-characterized epigenetic effects.


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.


Cell Metabolism | 2013

mTOR Complex 2 Controls Glycolytic Metabolism in Glioblastoma through FoxO Acetylation and Upregulation of c-Myc

Kenta Masui; Kazuhiro Tanaka; David Akhavan; Ivan Babic; Beatrice Gini; Tomoo Matsutani; Akio Iwanami; Feng Liu; Genaro R. Villa; Yuchao Gu; Carl Campos; Shaojun Zhu; Huijun Yang; William H. Yong; Timothy F. Cloughesy; Ingo K. Mellinghoff; Webster K. Cavenee; Reuben J. Shaw; Paul S. Mischel

Aerobic glycolysis (the Warburg effect) is a core hallmark of cancer, but the molecular mechanisms underlying it remain unclear. Here, we identify an unexpected central role for mTORC2 in cancer metabolic reprogramming where it controls glycolytic metabolism by ultimately regulating the cellular level of c-Myc. We show that mTORC2 promotes inactivating phosphorylation of class IIa histone deacetylases, which leads to the acetylation of FoxO1 and FoxO3, and this in turn releases c-Myc from a suppressive miR-34c-dependent network. These central features of activated mTORC2 signaling, acetylated FoxO, and c-Myc levels are highly intercorrelated in clinical samples and with shorter survival of GBM patients. These results identify a specific, Akt-independent role for mTORC2 in regulating glycolytic metabolism in cancer.


Science Translational Medicine | 2015

Glutamine-based PET imaging facilitates enhanced metabolic evaluation of gliomas in vivo

Sriram Venneti; Mark Dunphy; Hanwen Zhang; Kenneth L. Pitter; Patrick Zanzonico; Carl Campos; Sean Carlin; Gaspare La Rocca; Serge K. Lyashchenko; Karl Ploessl; Daniel Rohle; Antonio Omuro; Justin R. Cross; Cameron Brennan; Wolfgang A. Weber; Eric C. Holland; Ingo K. Mellinghoff; Hank F. Kung; Jason S. Lewis; Craig B. Thompson

Glutamine-based PET imaging takes advantage of gliomas’ glutamine addiction and can be used to assess metabolic nutrient uptake in gliomas. A PET approach to brain tumors Positron emission tomography, or PET, is a common method of imaging tumors by detecting their uptake of a radioactively labeled tracer. Radiolabeled glucose, in particular, is often used for this type of imaging, because tumor cells are often highly dependent on glycolysis and require large amounts of glucose to maintain their metabolism. Unfortunately, this method cannot be used to image brain tumors, because regular brain cells are also highly dependent on glucose. Now, Venneti et al. have used mouse models and human patients to show that radiolabeled glutamine, which is also taken up by tumor cells, can be used to image brain tumors and distinguish them from normal brain and even from tumors that are no longer growing. Glucose and glutamine are the two principal nutrients that cancer cells use to proliferate and survive. Many cancers show altered glucose metabolism, which constitutes the basis for in vivo positron emission tomography (PET) imaging with 18F-fluorodeoxyglucose (18F-FDG). However, 18F-FDG is ineffective in evaluating gliomas because of high background uptake in the brain. Glutamine metabolism is also altered in many cancers, and we demonstrate that PET imaging in vivo with the glutamine analog 4-18F-(2S,4R)-fluoroglutamine (18F-FGln) shows high uptake in gliomas but low background brain uptake, facilitating clear tumor delineation. Chemo/radiation therapy reduced 18F-FGln tumor avidity, corresponding with decreased tumor burden. 18F-FGln uptake was not observed in animals with a permeable blood-brain barrier or neuroinflammation. We translated these findings to human subjects, where 18F-FGln showed high tumor/background ratios with minimal uptake in the surrounding brain in human glioma patients with progressive disease. These data suggest that 18F-FGln is avidly taken up by gliomas, can be used to assess metabolic nutrient uptake in gliomas in vivo, and may serve as a valuable tool in the clinical management of gliomas.


Clinical Cancer Research | 2012

IDH Mutation and Neuroglial Developmental Features Define Clinically Distinct Subclasses of Lower Grade Diffuse Astrocytic Glioma

Daniel Gorovets; Kasthuri Kannan; Ronglai Shen; Edward R. Kastenhuber; Nasrin Islamdoust; Carl Campos; Elena Pentsova; Adriana Heguy; Suresh C. Jhanwar; Ingo K. Mellinghoff; Timothy A. Chan; Jason T. Huse

Purpose: Diffuse gliomas represent the most prevalent class of primary brain tumor. Despite significant recent advances in the understanding of glioblastoma [World Health Organization (WHO) IV], its most malignant subtype, lower grade (WHO II and III) glioma variants remain comparatively understudied, especially in light of their notable clinical heterogeneity. Accordingly, we sought to identify and characterize clinically relevant molecular subclasses of lower grade diffuse astrocytic gliomas. Experimental Design: We conducted multidimensional molecular profiling, including global transcriptional analysis, on 101 lower grade diffuse astrocytic gliomas collected at our own institution and validated our findings using publically available gene expression and copy number data from large independent patient cohorts. Results: We found that IDH mutational status delineated molecularly and clinically distinct glioma subsets, with IDH mutant (IDH mt) tumors exhibiting TP53 mutations, platelet—derived growth factor receptor (PDGFR)A overexpression, and prolonged survival, and IDH wild-type (IDH wt) tumors exhibiting EGFR amplification, PTEN loss, and unfavorable disease outcome. Furthermore, global expression profiling revealed three robust molecular subclasses within lower grade diffuse astrocytic gliomas, two of which were predominantly IDH mt and one almost entirely IDH wt. IDH mt subclasses were distinguished from each other on the basis of TP53 mutations, DNA copy number abnormalities, and links to distinct stages of neurogenesis in the subventricular zone. This latter finding implicates discrete pools of neuroglial progenitors as cells of origin for the different subclasses of IDH mt tumors. Conclusion: We have elucidated molecularly distinct subclasses of lower grade diffuse astrocytic glioma that dictate clinical behavior and show fundamental associations with both IDH mutational status and neuroglial developmental stage. Clin Cancer Res; 18(9); 2490–501. ©2012 AACR.


Neuro-oncology | 2016

Integration of 2-hydroxyglutarate-proton magnetic resonance spectroscopy into clinical practice for disease monitoring in isocitrate dehydrogenase-mutant glioma

Mac Arena I De La Fuente; Robert J. Young; Jennifer Rubel; Marc K. Rosenblum; Jamie Tisnado; Samuel Briggs; Julio Arevalo-Perez; Justin R. Cross; Carl Campos; Kimberly Straley; Dongwei Zhu; Chuanhui Dong; Alissa A. Thomas; Antonio Omuro; Craig Nolan; Elena Pentsova; Thomas Kaley; Jung H. Oh; Ralph Noeske; Elizabeth A. Maher; Changho Choi; Philip H. Gutin; Andrei I. Holodny; Katharine Yen; Lisa M. DeAngelis; Ingo K. Mellinghoff; Sunitha B. Thakur

BACKGROUND The majority of WHO grades II and III gliomas harbor a missense mutation in the metabolic gene isocitrate dehydrogenase (IDH) and accumulate the metabolite R-2-hydroxyglutarate (R-2HG). Prior studies showed that this metabolite can be detected in vivo using proton magnetic-resonance spectroscopy (MRS), but the sensitivity of this methodology and its clinical implications are unknown. METHODS We developed an MR imaging protocol to integrate 2HG-MRS into routine clinical glioma imaging and examined its performance in 89 consecutive glioma patients. RESULTS Detection of 2-hydroxyglutarate (2HG) in IDH-mutant gliomas was closely linked to tumor volume, with sensitivity ranging from 8% for small tumors (<3.4 mL) to 91% for larger tumors (>8 mL). In patients undergoing 2HG-MRS prior to surgery, tumor levels of 2HG corresponded with tumor cellularity but not with tumor grade or mitotic index. Cytoreductive therapy resulted in a gradual decrease in 2HG levels with kinetics that closely mirrored changes in tumor volume. CONCLUSIONS Our study demonstrates that 2HG-MRS can be linked with routine MR imaging to provide quantitative measurements of 2HG in glioma and may be useful as an imaging biomarker to monitor the abundance of IDH-mutant tumor cells noninvasively during glioma therapy and disease monitoring.


Cancer Discovery | 2017

Ibrutinib Unmasks Critical Role of Bruton Tyrosine Kinase in Primary CNS Lymphoma

Christian Grommes; Alessandro Pastore; Nicolaos Palaskas; Sarah S Tang; Carl Campos; Derrek Schartz; Paolo Codega; Donna Nichol; Owen Clark; Wan-Ying Hsieh; Daniel Rohle; Marc K. Rosenblum; Agnes Viale; Viviane Tabar; Cameron Brennan; Igor T. Gavrilovic; Thomas Kaley; Craig Nolan; Antonio Omuro; Elena Pentsova; Alissa A. Thomas; Elina Tsyvkin; Ariela Noy; M. Lia Palomba; Paul A. Hamlin; Craig S. Sauter; Craig H. Moskowitz; Julia Wolfe; Ahmet Dogan; Minhee Won

Bruton tyrosine kinase (BTK) links the B-cell antigen receptor (BCR) and Toll-like receptors with NF-κB. The role of BTK in primary central nervous system (CNS) lymphoma (PCNSL) is unknown. We performed a phase I clinical trial with ibrutinib, the first-in-class BTK inhibitor, for patients with relapsed or refractory CNS lymphoma. Clinical responses to ibrutinib occurred in 10 of 13 (77%) patients with PCNSL, including five complete responses. The only PCNSL with complete ibrutinib resistance harbored a mutation within the coiled-coil domain of CARD11, a known ibrutinib resistance mechanism. Incomplete tumor responses were associated with mutations in the B-cell antigen receptor-associated protein CD79B. CD79B-mutant PCNSLs showed enrichment of mammalian target of rapamycin (mTOR)-related gene sets and increased staining with PI3K/mTOR activation markers. Inhibition of the PI3K isoforms p110α/p110δ or mTOR synergized with ibrutinib to induce cell death in CD79B-mutant PCNSL cells.Significance: Ibrutinib has substantial activity in patients with relapsed or refractory B-cell lymphoma of the CNS. Response rates in PCNSL were considerably higher than reported for diffuse large B-cell lymphoma outside the CNS, suggesting a divergent molecular pathogenesis. Combined inhibition of BTK and PI3K/mTOR may augment the ibrutinib response in CD79B-mutant human PCNSLs. Cancer Discov; 7(9); 1018-29. ©2017 AACR.See related commentary by Lakshmanan and Byrd, p. 940This article is highlighted in the In This Issue feature, p. 920.


eLife | 2014

A kinase-independent function of AKT promotes cancer cell survival

Igor Vivanco; Zhi C Chen; Barbara E. Tanos; Barbara Oldrini; Wan-Ying Hsieh; Nicolas Yannuzzi; Carl Campos; Ingo K. Mellinghoff

The serine–threonine kinase AKT regulates proliferation and survival by phosphorylating a network of protein substrates. In this study, we describe a kinase-independent function of AKT. In cancer cells harboring gain-of-function alterations in MET, HER2, or Phosphatidyl-Inositol-3-Kinase (PI3K), catalytically inactive AKT (K179M) protected from drug induced cell death in a PH-domain dependent manner. An AKT kinase domain mutant found in human melanoma (G161V) lacked enzymatic activity in vitro and in AKT1/AKT2 double knockout cells, but promoted growth factor independent survival of primary human melanocytes. ATP-competitive AKT inhibitors failed to block the kinase-independent function of AKT, a liability that limits their effectiveness compared to allosteric AKT inhibitors. Our results broaden the current view of AKT function and have important implications for the development of AKT inhibitors for cancer. DOI: http://dx.doi.org/10.7554/eLife.03751.001


The Journal of Nuclear Medicine | 2013

Imaging Tumor Burden in the Brain with 89Zr-Transferrin

Michael J. Evans; Jason P. Holland; Samuel L. Rice; Michael G. Doran; Sarah M. Cheal; Carl Campos; Sean Carlin; Ingo K. Mellinghoff; Charles L. Sawyers; Jason S. Lewis

A noninvasive technology that indiscriminately detects tumor tissue in the brain could substantially enhance the management of primary or metastatic brain tumors. Although the documented molecular heterogeneity of diseases that initiate or eventually deposit in the brain may preclude identifying a single smoking-gun molecular biomarker, many classes of brain tumors are generally avid for transferrin. Therefore, we reasoned that applying a radiolabeled derivative of transferrin (89Zr-labeled transferrin) may be an effective strategy to more thoroughly identify tumor tissue in the brain, regardless of the tumor’s genetic background. Methods: Transferrin was radiolabeled with 89Zr, and its properties with respect to human models of glioblastoma multiforme were studied in vivo. Results: In this report, we show proof of concept that 89Zr-labeled transferrin (89Zr-transferrin) localizes to genetically diverse models of glioblastoma multiforme in vivo. Moreover, we demonstrate that 89Zr-transferrin can detect an orthotopic lesion with exceptional contrast. Finally, the tumor-to-brain contrast conferred by 89Zr-transferrin vastly exceeded that observed with 18F-FDG, currently the most widely used radiotracer to assess tumor burden in the brain. Conclusion: The results from this study suggest that 89Zr-transferrin could be a broadly applicable tool for identifying and monitoring tumors in the brain, with realistic potential for near-term clinical translation.

Collaboration


Dive into the Carl Campos's collaboration.

Top Co-Authors

Avatar

Ingo K. Mellinghoff

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Cameron Brennan

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Daniel Rohle

Memorial Sloan Kettering Cancer Center

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

Antonio Omuro

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Christian Grommes

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Igor Vivanco

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Marc K. Rosenblum

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Sean Carlin

Memorial Sloan Kettering Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge