Network


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

Hotspot


Dive into the research topics where Diego Carrera is active.

Publication


Featured researches published by Diego Carrera.


Journal of Clinical Investigation | 2017

Isocitrate dehydrogenase mutations suppress STAT1 and CD8 + T cell accumulation in gliomas

Gary Kohanbash; Diego Carrera; Shruti Shrivastav; Brian Ahn; Naznin Jahan; Tali Mazor; Zinal Chheda; Kira M. Downey; Payal Watchmaker; Casey Beppler; Rolf Warta; Nduka A. Amankulor; Christel Herold-Mende; Joseph F. Costello; Hideho Okada

Mutations in the isocitrate dehydrogenase genes IDH1 and IDH2 are among the first genetic alterations observed during the development of lower-grade glioma (LGG). LGG-associated IDH mutations confer gain-of-function activity by converting &agr;-ketoglutarate to the oncometabolite R-2-hydroxyglutarate (2HG). Clinical samples and gene expression data from The Cancer Genome Atlas (TCGA) demonstrate reduced expression of cytotoxic T lymphocyte–associated genes and IFN-&ggr;–inducible chemokines, including CXCL10, in IDH-mutated (IDH-MUT) tumors compared with IDH-WT tumors. Given these findings, we have investigated the impact of IDH mutations on the immunological milieu in LGG. In immortalized normal human astrocytes (NHAs) and syngeneic mouse glioma models, the introduction of mutant IDH1 or treatment with 2HG reduced levels of CXCL10, which was associated with decreased production of STAT1, a regulator of CXCL10. Expression of mutant IDH1 also suppressed the accumulation of T cells in tumor sites. Reductions in CXCL10 and T cell accumulation were reversed by IDH-C35, a specific inhibitor of mutant IDH1. Furthermore, IDH-C35 enhanced the efficacy of vaccine immunotherapy in mice bearing IDH-MUT gliomas. Our findings demonstrate a mechanism of immune evasion in IDH-MUT gliomas and suggest that specific inhibitors of mutant IDH may improve the efficacy of immunotherapy in patients with IDH-MUT gliomas.


Journal of Experimental Medicine | 2018

Novel and shared neoantigen derived from histone 3 variant H3.3K27M mutation for glioma T cell therapy

Zinal Chheda; Gary Kohanbash; Kaori Okada; Naznin Jahan; John Sidney; Matteo Pecoraro; Xinbo Yang; Diego Carrera; Kira M. Downey; Shruti Shrivastav; Shuming Liu; Yi Lin; Chetana Lagisetti; Pavlina Chuntova; Payal Watchmaker; Sabine Mueller; Ian F. Pollack; Raja Rajalingam; Angel M. Carcaboso; Matthias Mann; Alessandro Sette; K. Christopher Garcia; Yafei Hou; Hideho Okada

The median overall survival for children with diffuse intrinsic pontine glioma (DIPG) is less than one year. The majority of diffuse midline gliomas, including more than 70% of DIPGs, harbor an amino acid substitution from lysine (K) to methionine (M) at position 27 of histone 3 variant 3 (H3.3). From a CD8+ T cell clone established by stimulation of HLA-A2+ CD8+ T cells with synthetic peptide encompassing the H3.3K27M mutation, complementary DNA for T cell receptor (TCR) &agr;- and &bgr;-chains were cloned into a retroviral vector. TCR-transduced HLA-A2+ T cells efficiently killed HLA-A2+H3.3K27M+ glioma cells in an antigen- and HLA-specific manner. Adoptive transfer of TCR-transduced T cells significantly suppressed the progression of glioma xenografts in mice. Alanine-scanning assays suggested the absence of known human proteins sharing the key amino acid residues required for recognition by the TCR, suggesting that the TCR could be safely used in patients. These data provide us with a strong basis for developing T cell–based therapy targeting this shared neoepitope.


Genome Biology | 2017

Single-cell profiling of human gliomas reveals macrophage ontogeny as a basis for regional differences in macrophage activation in the tumor microenvironment

Sören Müller; Gary Kohanbash; S. John Liu; Beatriz Alvarado; Diego Carrera; Aparna Bhaduri; Payal Watchmaker; Garima Yagnik; Elizabeth Di Lullo; Martina Malatesta; Nduka Amankulor; Arnold R. Kriegstein; Daniel A. Lim; Manish K. Aghi; Hideho Okada; Aaron Diaz

BackgroundTumor-associated macrophages (TAMs) are abundant in gliomas and immunosuppressive TAMs are a barrier to emerging immunotherapies. It is unknown to what extent macrophages derived from peripheral blood adopt the phenotype of brain-resident microglia in pre-treatment gliomas. The relative proportions of blood-derived macrophages and microglia have been poorly quantified in clinical samples due to a paucity of markers that distinguish these cell types in malignant tissue.ResultsWe perform single-cell RNA-sequencing of human gliomas and identify phenotypic differences in TAMs of distinct lineages. We isolate TAMs from patient biopsies and compare them with macrophages from non-malignant human tissue, glioma atlases, and murine glioma models. We present a novel signature that distinguishes TAMs by ontogeny in human gliomas. Blood-derived TAMs upregulate immunosuppressive cytokines and show an altered metabolism compared to microglial TAMs. They are also enriched in perivascular and necrotic regions. The gene signature of blood-derived TAMs, but not microglial TAMs, correlates with significantly inferior survival in low-grade glioma. Surprisingly, TAMs frequently co-express canonical pro-inflammatory (M1) and alternatively activated (M2) genes in individual cells.ConclusionsWe conclude that blood-derived TAMs significantly infiltrate pre-treatment gliomas, to a degree that varies by glioma subtype and tumor compartment. Blood-derived TAMs do not universally conform to the phenotype of microglia, but preferentially express immunosuppressive cytokines and show an altered metabolism. Our results argue against status quo therapeutic strategies that target TAMs indiscriminately and in favor of strategies that specifically target immunosuppressive blood-derived TAMs.


JCI insight | 2017

GLUT3 upregulation promotes metabolic reprogramming associated with antiangiogenic therapy resistance

Ruby Kuang; Arman Jahangiri; Smita Mascharak; Alan Nguyen; Ankush Chandra; Patrick M. Flanigan; Garima Yagnik; Jeffrey Wagner; Michael De Lay; Diego Carrera; Brandyn Castro; Josie Hayes; Maxim Sidorov; Jose Luiz Izquierdo Garcia; Pia Eriksson; Sabrina M. Ronen; Joanna J. Phillips; Annette M. Molinaro; Suneil K. Koliwad; Manish K. Aghi

Clinical trials revealed limited response duration of glioblastomas to VEGF-neutralizing antibody bevacizumab. Thriving in the devascularized microenvironment occurring after antiangiogenic therapy requires tumor cell adaptation to decreased glucose, with 50% less glucose identified in bevacizumab-treated xenografts. Compared with bevacizumab-responsive xenograft cells, resistant cells exhibited increased glucose uptake, glycolysis, 13C NMR pyruvate to lactate conversion, and survival in low glucose. Glucose transporter 3 (GLUT3) was upregulated in bevacizumab-resistant versus sensitive xenografts and patient specimens in a HIF-1α-dependent manner. Resistant versus sensitive cell mitochondria in oxidative phosphorylation-selective conditions produced less ATP. Despite unchanged mitochondrial numbers, normoxic resistant cells had lower mitochondrial membrane potential than sensitive cells, confirming poorer mitochondrial health, but avoided the mitochondrial dysfunction of hypoxic sensitive cells. Thin-layer chromatography revealed increased triglycerides in bevacizumab-resistant versus sensitive xenografts, a change driven by mitochondrial stress. A glycogen synthase kinase-3β inhibitor suppressing GLUT3 transcription caused greater cell death in bevacizumab-resistant than -responsive cells. Overexpressing GLUT3 in tumor cells recapitulated bevacizumab-resistant cell features: survival and proliferation in low glucose, increased glycolysis, impaired oxidative phosphorylation, and rapid in vivo proliferation only slowed by bevacizumab to that of untreated bevacizumab-responsive tumors. Targeting GLUT3 or the increased glycolysis reliance in resistant tumors could unlock the potential of antiangiogenic treatments.


Cancer Research | 2017

Abstract 3767: Identification of a novel and a shared H3.3K27M mutation derived neoantigen epitope and H3.3K27M specific TCR engineered T cell therapy for glioma

Zinal Chheda; Gary Kohanbash; John Sidney; Kaori Okada; Naznin Jahan; Diego Carrera; Payal Watchmaker; Kira M. Downey; Shuming Liu; Shruti Shrivastav; Sabine Mueller; Ian F. Pollack; Angel M. Carcaboso; Alessandro Sette; Yafei Hou; Hideho Okada

Brain cancers are the leading cause of cancer related mortality in children and young adults with median overall survival of 9-10 months and hence represents a significant unmet medical need. Genome-wide sequencing efforts of pediatric gliomas have identified a recurrent and shared missense mutation in the gene encoding the replication-independent variant of histone 3, H3.3. Approximately 70% of diffuse intrinsic pontine gliomas (DIPG) and 50% of thalamic and other midline gliomas harbor the amino acid substitution from lysine (K) to methionine (M) at the position 27 of H3.3 gene. Tumor specific missense mutations are not subjected to self-tolerance and can be suitable targets (neoantigens) for cancer immunotherapy. Herein, we evaluated whether the H3.3K27M mutation can induce specific cytotoxic T lymphocyte (CTL) response in HLA-A2+ T cells. In vitro stimulation of HLA-A2+ donor derived CD8+ T cells with a synthetic peptide encompassing the H3.3K27M mutation (H3.3K27M epitope) induced CTL lines which recognized not only T2 cells loaded with the synthetic H3.3K27M epitope peptide but also lysed the HLA-A2+ DIPG cells which endogenously harbor the H3.3K27M mutation. On the other hand, the CTL lines did not react to either HLA-A2+ but H3.3K27M- negative DIPG cell lines or H3.3K27M positive but HLA-A2 negative DIPG cells. The H3.3K27M epitope peptide but not the non-mutant counterpart indicated an excellent binding affinity (Kd 151nM) to HLA-A2 based on competitive binding inhibition assay. From CTL clones with high and specific affinities to HLA-A2-H3.3K27M-tetramer, cDNAs for T cell receptor (TCR) alpha and beta chains were cloned into a retroviral vector. Human HLA-A2+ T cells transduced with the TCR demonstrated antigen specific reactivity as well as anti-glioma responses in vitro. Peptide titration assays suggested that the H3.3K27M specific TCR had the half-maximal reactivity for peptide recognition of around 100nM. Furthermore, critically important for safety of clinical application, alanine scanning demonstrated that the key amino acid sequence motif in the epitope of the TCR reactivity is not shared by any known human protein. Finally, intravenous administration of T cells transduced with H3.3K27M specific TCR significantly inhibited the growth of intracranial HLA-A2+ H3.3K27M positive glioma xenografts in immune deficient NSG mice. These data provide us with a strong basis for developing peptide based vaccines as well as adoptive transfer therapy using autologous T cells transduced with the H3.3K27M specific TCR. Acknowledgements: This study is supported by the NIH/NINDS (1RO1NS096954), V Foundation and Parker Institution for Cancer Immunotherapy. Citation Format: Zinal Chheda, Gary Kohanbash, John Sidney, Kaori Okada, Naznin Jahan, Diego Carrera, Payal Watchmaker, Kira Downey, Shuming Liu, Shruti Shrivastav, Sabine Mueller, Ian F. Pollack, Angel M. Carcaboso, Alessandro Sette, Yafei Hou, Hideho Okada. Identification of a novel and a shared H3.3K27M mutation derived neoantigen epitope and H3.3K27M specific TCR engineered T cell therapy for glioma [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 3767. doi:10.1158/1538-7445.AM2017-3767


Neurosurgical Focus | 2018

Disparities in health care determine prognosis in newly diagnosed glioblastoma

Ankush Chandra; Jonathan Rick; Cecilia L. Dalle Ore; Darryl Lau; Alan T. Nguyen; Diego Carrera; Alexander Bonte; Annette M. Molinaro; Philip V. Theodosopoulos; Michael W. McDermott; Mitchel S. Berger; Manish K. Aghi


Neuro-oncology | 2017

IMMU-41. H3.3K27M MUTATION-DERIVED NOVEL NEOANTIGEN – CHARACTERIZATION OF THE HLA-A2-BINDING EPITOPE AND A SPECIFIC T CELL RECEPTOR FOR DEVELOPMENT OF T CELL-BASED IMMUNOTHERAPY

Zinal Chheda; Gary Kohanbash; Kaori Okada; Naznin Jahan; John Sidney; Matteo Pecoraro; Diego Carrera; Shruti Shrivastav; Shuming Liu; Kira M. Downey; Pavlina Chuntova; Payal Watchmaker; Sabine Mueller; Ian F. Pollack; Raja Rajalingam; Angel M. Carcaboso; Matthias Mann; Alessandro Sette; Yafei Hou; Hideho Okada


Neuro-oncology | 2017

TMIC-11. TUMOR GENETICS AND MACROPHAGE ONTOGENY SHAPE THE INNATE IMMUNE RESPONSE TO GLIOMA

Soeren Mueller; Gary Kohanbash; Siyuan Liu; Diego Carrera; Payal Watchmaker; Naznin Jahan; Beatriz Alvarado; Dan Lim; Manish K. Aghi; Hideho Okada; Aaron Diaz


Neuro-oncology | 2017

TMOD-12. AN ONCOLYTIC MEASLES VIRUS SENSITIVE GROUP 3 MEDULLOBLASTOMA MODEL IN IMMUNE-COMPETENT MICE

Sangeet Lal; Diego Carrera; Joanna J. Phillips; William A. Weiss; Corey Raffel


Neuro-oncology | 2017

IMMU-52. SELECTION OF GLIOMA T-CELL THERAPY TARGETS BASED ON THE ANALYSIS OF TUMOR IMMUNOPEPTIDOME AND EXPRESSION PROFILES

Diego Carrera; Josie Hayes; Soeren Mueller; Samuel Shelton; Toni Weinschenk; Colette Song; Jens Fritsche; Oliver Schoor; Sabrina Kuttruff-Coqui; Norbert Hilf; Gary Kohanbash; Payal Watchmaker; Michael C. Oldham; Joseph F. Costello; Harpreet Singh; Hideho Okada

Collaboration


Dive into the Diego Carrera's collaboration.

Top Co-Authors

Avatar

Gary Kohanbash

University of California

View shared research outputs
Top Co-Authors

Avatar

Hideho Okada

University of California

View shared research outputs
Top Co-Authors

Avatar

Naznin Jahan

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kira M. Downey

University of California

View shared research outputs
Top Co-Authors

Avatar

Manish K. Aghi

University of California

View shared research outputs
Top Co-Authors

Avatar

Zinal Chheda

University of California

View shared research outputs
Top Co-Authors

Avatar

Alessandro Sette

La Jolla Institute for Allergy and Immunology

View shared research outputs
Top Co-Authors

Avatar

Ian F. Pollack

Boston Children's Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge