Network


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

Hotspot


Dive into the research topics where Shruti Shrivastav is active.

Publication


Featured researches published by Shruti Shrivastav.


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.


Oncogene | 2017

Macrophage migration inhibitory factor downregulation: a novel mechanism of resistance to anti-angiogenic therapy

Brandyn Castro; Patrick M. Flanigan; Arman Jahangiri; D Hoffman; William Chen; Ruby Kuang; M De Lay; Garima Yagnik; Jeffrey Wagner; Smita Mascharak; Maxim Sidorov; Shruti Shrivastav; Gary Kohanbash; Hideho Okada; Manish K. Aghi

Anti-angiogenic therapies for cancer such as VEGF neutralizing antibody bevacizumab have limited durability. While mechanisms of resistance remain undefined, it is likely that acquired resistance to anti-angiogenic therapy will involve alterations of the tumor microenvironment. We confirmed increased tumor-associated macrophages in bevacizumab-resistant glioblastoma patient specimens and two novel glioblastoma xenograft models of bevacizumab resistance. Microarray analysis suggested downregulated macrophage migration inhibitory factor (MIF) to be the most pertinent mediator of increased macrophages. Bevacizumab-resistant patient glioblastomas and both novel xenograft models of resistance had less MIF than bevacizumab-naive tumors, and harbored more M2/protumoral macrophages that specifically localized to the tumor edge. Xenografts expressing MIF-shRNA grew more rapidly with greater angiogenesis and had macrophages localizing to the tumor edge which were more prevalent and proliferative, and displayed M2 polarization, whereas bevacizumab-resistant xenografts transduced to upregulate MIF exhibited the opposite changes. Bone marrow-derived macrophage were polarized to an M2 phenotype in the presence of condition-media derived from bevacizumab-resistant xenograft-derived cells, while recombinant MIF drove M1 polarization. Media from macrophages exposed to bevacizumab-resistant tumor cell conditioned media increased glioma cell proliferation compared with media from macrophages exposed to bevacizumab-responsive tumor cell media, suggesting that macrophage polarization in bevacizumab-resistant xenografts is the source of their aggressive biology and results from a secreted factor. Two mechanisms of bevacizumab-induced MIF reduction were identified: (1) bevacizumab bound MIF and blocked MIF-induced M1 polarization of macrophages; and (2) VEGF increased glioma MIF production in a VEGFR2-dependent manner, suggesting that bevacizumab-induced VEGF depletion would downregulate MIF. Site-directed biopsies revealed enriched MIF and VEGF at the enhancing edge in bevacizumab-naive patients. This MIF enrichment was lost in bevacizumab-resistant glioblastomas, driving a tumor edge M1-to-M2 transition. Thus, bevacizumab resistance is driven by reduced MIF at the tumor edge causing proliferative expansion of M2 macrophages, which in turn promotes tumor growth.


Journal of Neuro-oncology | 2016

Expression and prognostic impact of immune modulatory molecule PD-L1 in meningioma

Seunggu J. Han; Gerald F. Reis; Gary Kohanbash; Shruti Shrivastav; Stephen T. Magill; Annette M. Molinaro; Michael W. McDermott; Philip V. Theodosopoulos; Manish K. Aghi; Mitchel S. Berger; Nicholas Butowski; Igor J. Barani; Joanna J. Phillips; Arie Perry; Hideho Okada

While immunotherapy may offer promising new approaches for high grade meningiomas, little is currently known of the immune landscape in meningiomas. We sought to characterize the immune microenvironment and a potentially targetable antigen mesothelin across WHO grade I-III cases of meningiomas, and how infiltrating immune populations relate to patient outcomes. Immunohistochemistry was performed on tissue microarrays constructed from 96 meningioma cases. The cohort included 16 WHO grade I, 62 WHO grade II, and 18 WHO grade III tumors. Immunohistochemistry was performed using antibodies against CD3, CD8, CD20, CD68, PD-L1, and mesothelin. Dual staining using anti-PD-L1 and anti-CD68 antibodies was performed, and automated cell detection and positive staining detection algorithms were utilized. Greater degree of PD-L1 expression was found in higher grade tumors. More specifically, higher grade tumors contained increased numbers of intratumoral CD68−, PD-L1+ cells (p = 0.022), but did not contain higher numbers of infiltrating CD68+, PD-L1+ cells (p = 0.30). Higher PD-L1+/CD68− expression was independently predictive of worse overall survival in our cohort when accounting for grade, performance status, extent of resection, and recurrence history (p = 0.014). Higher expression of PD-L1+/CD68− was also present in tumors that had undergone prior radiotherapy (p = 0.024). Approximately quarter of meningiomas overexpressed mesothelin to levels equivalent to those found in pancreatic carcinomas and malignant mesotheliomas. The association with poor survival outcomes in our study suggests that PD-L1 may play a significant biologic role in the aggressive phenotype of higher grade meningiomas. Thus, immunotherapeutic strategies such as checkpoint inhibition may have clinical utility in PD-L1 overexpressing meningiomas.


Journal for ImmunoTherapy of Cancer | 2015

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

Yafei Hou; Gary Kohanbash; Kaori Okada; Shruti Shrivastav; Matthew Smith-Cohn; Theodore Nicolaides; Sabine Mueller; Angel M. Carcaboso; Ian F. Pollack; Hideho Okada

Meeting abstracts Malignant gliomas, such as glioblastoma (GBM) and diffuse intrinsic pontine gliomas (DIPG), are lethal brain tumors in both adults and children. Indeed, brain tumors are the leading cause of cancer-related mortality and morbidity in children. Children with DIPG have one-year


Cancer immunology research | 2016

Abstract A117: Novel and shared neoantigen for glioma T cell therapy derived from histone 3 variant H3.3 K27M mutation

Yafei Hou; Gary Kohanbash; Kaori Okada; Shruti Shrivastav; Matthew Smith-Cohn; Theodore Nicolaides; Sabine Mueller; Angel M. Carcaboso; Ian F. Pollack; Hideho Okada

Malignant gliomas, such as glioblastoma (GBM) and diffuse intrinsic pontine gliomas (DIPG), are lethal brain tumors in both adults and children. Indeed, brain tumors are the leading cause of cancer-related mortality and morbidity in children. Children with DIPG have one-year progression-free survival rates below 25%, and median overall survival of 9 to 10 months with current treatment. Recent genetic studies have revealed that malignant gliomas in children and young adults often show shared missense mutations, which encodes the replication-independent histone 3 variant H3.3. Approximately 30 % of overall GBM and over 70% of DIPG cases harbor the amino-acid substitution from lysine (K) to methionine (M) at the position 27 of H3.3. The H3.3 K27M mutation in DIPG is universally associated with shorter survival compared with patients with non-mutated H3.3. T-cells of the adaptive immune system are normally tolerant to wild-type self-proteins, but can recognize mutated peptide epitopes as non-self. Hence, tumor-specific missense mutations can be suitable targets (i.e. neoantigens) for cancer immunotherapy, such as cancer vaccines and adoptive T cell transfer therapy. We evaluated whether H3.3-derived peptides that encompass the H3.3 K27M mutation can induce specific cytotoxic T lymphocyte (CTL) responses in human leukocyte antigen (HLA)-A2+ CD8+ T-cells. For prediction of HLA-A2-binding epitopes, an algorithm integrating peptide binding to HLA (NetMHC 3.4 server) and a proteosomal cleavage site prediction system (http://paproc.de/) was used. Four candidate peptides encompassing different amino-acid positions around the H3.3 K27M mutation were synthesized, and peptide-specific CTL lines and clones were generated from peripheral blood mononuclear cells of HLA-A2+ donors by in vitro stimulation with each of the synthetic peptides. One of the 4 peptides (the H3.3.K27M epitope, hereafter) induced CTL lines which recognized not only the synthetic peptide loaded on T2 cells but also lysed HLA-A2+ DIPG cell lines which endogenously harbor the H3.3.K27M mutation. On the other hand, CTL lines did not react to HLA-A2+, H3.3 K27M mutation-negative cells or HLA-A2-negative, H3.3 K27M mutation+ cells. Furthermore, CTL clones with high and specific affinities to HLA-A2-H3.3.K27M-tetramer were successfully obtained, and α- and β-chain cDNAs from high-affinity T cell receptors (TCR)s were cloned into a lentiviral vector. Additional studies are underway to determine antigen specificity, key epitope residues in the epitope and possible cross-reactivity to naturally existing variants using T-cells transduced with the lentiviral vector encoding the TCR. Assessments of in vivo immune responses to the epitope peptide and preclinical confirmation for absence of autoimmunity are also underway using HLA-A2-transgenic 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 TCR. Our experience with conducting immunotherapy trials in these patients will facilitate the translation. Citation Format: Yafei Hou, Gary Kohanbash, Kaori Okada, Shruti Shrivastav, Matthew Smith-Cohn, Theodore Nicolaides, Sabine Mueller, Angel Montero Carcaboso, Ian F. Pollack, Hideho Okada. Novel and shared neoantigen for glioma T cell therapy derived from histone 3 variant H3.3 K27M mutation. [abstract]. In: Proceedings of the CRI-CIMT-EATI-AACR Inaugural International Cancer Immunotherapy Conference: Translating Science into Survival; September 16-19, 2015; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2016;4(1 Suppl):Abstract nr A117.


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


Journal for ImmunoTherapy of Cancer | 2015

IDH mutation-induced suppression of type-1 anti-glioma immune response

Gary Kohanbash; Shruti Shrivastav; Brian Ahn; Yafei Hou; Joseph F. Costello; Hideho Okada

Isocitrate dehydrogenase (IDH) mutations are the first mutations that occur during the oncogenic process of lower-grade glioma (LGG) and confers a novel gain-of-function activity by converting α-ketoglutarate (αKG) to 2-hydroxyglutarate (2HG), promoting DNA hyper-methylation. Our analysis of LGG cases from The Cancer Genome Atlas (TCGA) database revealed that IDH-mutant (IDH-Mut) cases exhibit decreased expression of type-1 effector T cell response-related genes, which are critical for anti-glioma immunity, including: CD8A, IFNG, OAS2, GZMA, EOMES, CXCL9 and CXCL10, compared with IDH-wild type (IDH-WT) cases. On the other hand, type-2 and regulatory T cell response-related genes, such as IL5 and TGFB1, are not significantly different between IDH-Mut vs. WT cases, indicating that the observed down-regulation of type-1 response-related genes does not merely represent a possible global gene suppression. Furthermore, IDH-Mut cases exhibit increased CXCL10 promotor methylation compared with WT cases. We thus hypothesized that IDH mutation-mediated tumor intrinsic mechanisms occurring within glioma cells may inhibit anti-tumor immunity to promote tumor growth. In vitro, a normal human astrocyte (NHA) cell line transfected with IDH1-Mut cDNA expressed lower levels of CXCL10 compared to NHA cells transfected with WT IDH1. Consistently, C57Bl/6 mouse-syngeneic astrocyte and glioma cell lines transfected with IDH1-Mut expressed lower levels of CXCL10 gene and protein, compared to control cells transfected with IDH-WT, which was restored following 30 day treatment of the cells with the IDH1 inhibitor, IDH-C35. Furthermore, in vivo orthotopic IDH1-Mut gliomas at 21 days post-intracranial injection in syngeneic mice expressed lower levels of T cell chemokines CXCL9 and CXCL10 as determined by RT-PCR and ELISA and reduced infiltration of CD3+CD8+ T cells as determined by flow cytometry and quantitative immunohistochemistry compared with control IDH1-WT gliomas. Further, an in vitro migration assay demonstrated reduced migration of T cells towards culture supernatants from IDH1-Mut cell lines compared with control supernatants derived from IDH1-WT cells. Overall, our data demonstrate that IDH mutations in tumor cells lead to reduced T cell attracting chemokines and reduced T cell accumulation in gliomas. Our analyses of the TCGA 450K gene methylation database suggest that the suppressed expression of OAS2 and CXCL10 in IDH1-Mut cases is associated with hypermethylation of the promoter for these genes. Indeed, treatment of IDH-Mut cell lines with demethylating agent 5-Aza-CdR restored CXCL10 expression levels. Our data suggest that IDH inhibitors and demethylation agents may be used to enhance T cell recruitment to LGG in combination with T cell based immunotherapies.


Neuro-oncology | 2015

MTR-01BEVACIZUMAB-INDUCED MIF DEPLETION: A NOVEL RESISTANCE MECHANISM IN GLIOBLASTOMA

Brandyn Castro; Arman Jahangiri; Ruby Kuang; Michael De Lay; Gary Kohanbash; Garima Yagnik; Liane Miller; Shruti Shrivastav; Hideho Okada; Manish K. Aghi


Neuro-oncology | 2015

IMPS-30NOVEL AND SHARED NEOANTIGEN FOR GLIOMA T CELL THERAPY DERIVED FROM HISTONE 3 VARIANT H3.3 K27M MUTATION

Yafei Hou; Gary Kohanbash; Kaori Okada; Shruti Shrivastav; Matthew Smith-Cohn; Theodore Nicolaides; Sabine Mueller; Angel M. Carcaboso; Ian F. Pollack; Hideho Okada

Collaboration


Dive into the Shruti Shrivastav'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

Yafei Hou

University of California

View shared research outputs
Top Co-Authors

Avatar

Diego Carrera

University of California

View shared research outputs
Top Co-Authors

Avatar

Ian F. Pollack

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Kaori 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

Sabine Mueller

University of California

View shared research outputs
Top Co-Authors

Avatar

Kira M. Downey

University of California

View shared research outputs
Top Co-Authors

Avatar

Zinal Chheda

University of California

View shared research outputs
Researchain Logo
Decentralizing Knowledge