Network


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

Hotspot


Dive into the research topics where Renate Starr is active.

Publication


Featured researches published by Renate Starr.


Blood | 2013

T cells expressing CD123-specific chimeric antigen receptors exhibit specific cytolytic effector functions and antitumor effects against human acute myeloid leukemia.

Armen Mardiros; Cedric Dos Santos; Tinisha McDonald; Christine E. Brown; Xiuli Wang; L. Elizabeth Budde; Lauren Hoffman; Brenda Aguilar; Wen-Chung Chang; William Bretzlaff; Brenda Chang; Mahesh Jonnalagadda; Renate Starr; Julie R. Ostberg; Michael C. Jensen; Ravi Bhatia; Stephen J. Forman

Induction treatments for acute myeloid leukemia (AML) have remained largely unchanged for nearly 50 years, and AML remains a disease of poor prognosis. Allogeneic hematopoietic cell transplantation can achieve cures in select patients and highlights the susceptibility of AML to donor-derived immunotherapy. The interleukin-3 receptor α chain (CD123) has been identified as a potential immunotherapeutic target because it is overexpressed in AML compared with normal hematopoietic stem cells. Therefore, we developed 2 chimeric antigen receptors (CARs) containing a CD123-specific single-chain variable fragment, in combination with a CD28 costimulatory domain and CD3-ζ signaling domain, targeting different epitopes on CD123. CD123-CAR-redirected T cells mediated potent effector activity against CD123+ cell lines as well as primary AML patient samples. CD123 CAR T cells did not eliminate granulocyte/macrophage and erythroid colony formation in vitro. Additionally, T cells obtained from patients with active AML can be modified to express CD123 CARs and are able to lyse autologous AML blasts in vitro. Finally, CD123 CAR T cells exhibited antileukemic activity in vivo against a xenogeneic model of disseminated AML. These results suggest that CD123 CAR T cells are a promising immunotherapy for the treatment of high-risk AML.


Cancer Research | 2009

Recognition and Killing of Brain Tumor Stem-Like Initiating Cells by CD8+ Cytolytic T Cells

Christine E. Brown; Renate Starr; Catalina Martinez; Brenda Aguilar; Massimo D'Apuzzo; Ivan Todorov; Chu Chih Shih; Behnam Badie; Michael Hudecek; Stanley R. Riddell; Michael C. Jensen

Solid tumors contain a subset of stem-like cells that are resistant to the cytotoxic effects of chemotherapy/radiotherapy, but their susceptibility to cytolytic T lymphocyte (CTL) effector mechanisms has not been well characterized. Using a panel of early-passage human brain tumor stem/initiating cell (BTSC) lines derived from high-grade gliomas, we show that BTSCs are subject to immunologic recognition and elimination by CD8(+) CTLs. Compared with serum-differentiated CD133(low) tumor cells and established glioma cell lines, BTSCs are equivalent with respect to expression levels of HLA class I and ICAM-1, similar in their ability to trigger degranulation and cytokine synthesis by antigen-specific CTLs, and equally susceptible to perforin-dependent CTL-mediated cytolysis. BTSCs are also competent in the processing and presentation of antigens as evidenced by the killing of these cells by CTL when antigen is endogenously expressed. Moreover, we show that CTLs can eliminate all BTSCs with tumor-initiating activity in an antigen-specific manner in vivo. Current models predict that curative therapies for many cancers will require the elimination of the stem/initiating population, and these studies lay the foundation for developing immunotherapeutic approaches to eradicate this tumor population.


Clinical Cancer Research | 2012

Stem-like tumor-initiating cells isolated from IL13Rα2 expressing gliomas are targeted and killed by IL13-zetakine-redirected T Cells.

Christine E. Brown; Renate Starr; Brenda Aguilar; Andrew F. Shami; Catalina Martinez; Massimo D'Apuzzo; Michael E. Barish; Stephen J. Forman; Michael C. Jensen

Purpose: To evaluate IL13Rα2 as an immunotherapeutic target for eliminating glioma stem–like cancer initiating cells (GSC) of high-grade gliomas, with particular focus on the potential of genetically engineered IL13Rα2-specific primary human CD8+ CTLs (IL13-zetakine+ CTL) to target this therapeutically resistant glioma subpopulation. Experimental Design: A panel of low-passage GSC tumor sphere (TS) and serum-differentiated glioma lines were expanded from patient glioblastoma specimens. These glioblastoma lines were evaluated for expression of IL13Rα2 and for susceptibility to IL13-zetakine+ CTL-mediated killing in vitro and in vivo. Results: We observed that although glioma IL13Rα2 expression varies between patients, for IL13Rα2pos cases this antigen was detected on both GSCs and more differentiated tumor cell populations. IL13-zetakine+ CTL were capable of efficient recognition and killing of both IL13Rα2pos GSCs and IL13Rα2pos differentiated cells in vitro, as well as eliminating glioma-initiating activity in an orthotopic mouse tumor model. Furthermore, intracranial administration of IL13-zetakine+ CTL displayed robust antitumor activity against established IL13Rα2pos GSC TS-initiated orthotopic tumors in mice. Conclusions: Within IL13Rα2 expressing high-grade gliomas, this receptor is expressed by GSCs and differentiated tumor populations, rendering both targetable by IL13-zetakine+ CTLs. Thus, our results support the potential usefullness of IL13Rα2-directed immunotherapeutic approaches for eradicating therapeutically resistant GSC populations. Clin Cancer Res; 18(8); 2199–209. ©2012 AACR.


Molecular Cancer Therapeutics | 2010

Sorafenib Induces Growth Arrest and Apoptosis of Human Glioblastoma Cells through the Dephosphorylation of Signal Transducers and Activators of Transcription 3

Fan Yang; Christine E. Brown; Ralf Buettner; Michael Hedvat; Renate Starr; Anna Scuto; Anne Schroeder; Michael C. Jensen; Richard Jove

Glioblastoma is the most common type of primary brain tumor and is rapidly progressive with few treatment options. Here, we report that sorafenib (≤10 μmol/L) inhibited cell proliferation and induced apoptosis in two established cell lines (U87 and U251) and two primary cultures (PBT015 and PBT022) from human glioblastomas. The effects of sorafenib on these tumor cells were associated with inhibiting phosphorylated signal transducers and activators of transcription 3 (STAT3; Tyr705). Expression of a constitutively activated STAT3 mutant partially blocked the effects of sorafenib, consistent with a role for STAT3 inhibition in the response to sorafenib. Phosphorylated Janus-activated kinase (JAK)1 was inhibited in U87 and U251 cells, whereas phosphorylated JAK2 was inhibited in primary cultures. Sodium vanadate, a general inhibitor of protein tyrosine phosphatases, blocked the inhibition of phosphorylation of STAT3 (Tyr705) induced by sorafenib. These data indicate that the inhibition of STAT3 activity by sorafenib involves both the inhibition of upstream kinases (JAK1 and JAK2) of STAT3 and increased phosphatase activity. Phosphorylation of AKT was also reduced by sorafenib. In contrast, mitogen-activated protein kinases were not consistently inhibited by sorafenib in these cells. Two key cyclins (D and E) and the antiapoptotic protein Mcl-1 were downregulated by sorafenib in both cell lines and primary cultures. Our data suggest that inhibition of STAT3 signaling by sorafenib contributes to growth arrest and induction of apoptosis in glioblastoma cells. These findings provide a rationale for potential treatment of malignant gliomas with sorafenib. Mol Cancer Ther; 9(4); 953–62. ©2010 AACR.


Journal of Immunology | 2007

Tumor-Derived Chemokine MCP-1/CCL2 Is Sufficient for Mediating Tumor Tropism of Adoptively Transferred T Cells

Christine E. Brown; Reena Vishwanath; Brenda Aguilar; Renate Starr; Joseph Najbauer; Karen S. Aboody; Michael C. Jensen

To exert a therapeutic effect, adoptively transferred tumor-specific CTLs must traffic to sites of tumor burden, exit the circulation, and infiltrate the tumor microenvironment. In this study, we examine the ability of adoptively transferred human CTL to traffic to tumors with disparate chemokine secretion profiles independent of tumor Ag recognition. Using a combination of in vivo tumor tropism studies and in vitro biophotonic chemotaxis assays, we observed that cell lines derived from glioma, medulloblastoma, and renal cell carcinoma efficiently chemoattracted ex vivo-expanded primary human T cells. We compared the chemokines secreted by tumor cell lines with high chemotactic activity with those that failed to elicit T cell chemotaxis (Daudi lymphoma, 10HTB neuroblastoma, and A2058 melanoma cells) and found a correlation between tumor-derived production of MCP-1/CCL2 (≥10 ng/ml) and T cell chemotaxis. Chemokine immunodepletion studies confirmed that tumor-derived MCP-1 elicits effector T cell chemotaxis. Moreover, MCP-1 is sufficient for in vivo T cell tumor tropism as evidenced by the selective accumulation of i.v. administered firefly luciferase-expressing T cells in intracerebral xenografts of tumor transfectants secreting MCP-1. These studies suggest that the capacity of adoptively transferred T cells to home to tumors may be, in part, dictated by the species and amounts of tumor-derived chemokines, in particular MCP-1.


Molecular Therapy | 2015

Chimeric Antigen Receptors With Mutated IgG4 Fc Spacer Avoid Fc Receptor Binding and Improve T Cell Persistence and Antitumor Efficacy

Mahesh Jonnalagadda; Armen Mardiros; Ryan Urak; Xiuli Wang; Lauren Hoffman; Alyssa Bernanke; Wen-Chung Chang; William Bretzlaff; Renate Starr; Saul J. Priceman; Julie R. Ostberg; Stephen J. Forman; Christine E. Brown

The success of adoptive therapy using chimeric antigen receptor (CAR)-expressing T cells partly depends on optimal CAR design. CARs frequently incorporate a spacer/linker region based on the constant region of either IgG1 or IgG4 to connect extracellular ligand-binding with intracellular signaling domains. Here, we evaluated the potential for the IgG4-Fc linker to result in off-target interactions with Fc gamma receptors (FcγRs). As proof-of-principle, we focused on a CD19-specific scFv-IgG4-CD28-zeta CAR and found that, in contrast to CAR-negative cells, CAR+ T cells bound soluble FcγRs in vitro and did not engraft in NSG mice. We hypothesized that mutations to avoid FcγR binding would improve CAR+ T cell engraftment and antitumor efficacy. Thus, we generated CD19-specific CARs with IgG4-Fc spacers that had either been mutated at two sites (L235E; N297Q) within the CH2 region (CD19R(EQ)) or incorporated a CH2 deletion (CD19Rch2Δ). These mutations reduced binding to soluble FcγRs without altering the ability of the CAR to mediate antigen-specific lysis. Importantly, CD19R(EQ) and CD19Rch2Δ T cells exhibited improved persistence and more potent CD19-specific antilymphoma efficacy in NSG mice. Together, these studies suggest that optimal CAR function may require the elimination of cellular FcγR interactions to improve T cell persistence and antitumor responses.


PLOS ONE | 2013

Glioma IL13Rα2 is associated with mesenchymal signature gene expression and poor patient prognosis.

Christine E. Brown; Charles Warden; Renate Starr; Xutao Deng; Behnam Badie; Yate-Ching Yuan; Stephen J. Forman; Michael E. Barish

A major challenge for successful immunotherapy against glioma is the identification and characterization of validated targets. We have taken a bioinformatics approach towards understanding the biological context of IL-13 receptor α2 (IL13Rα2) expression in brain tumors, and its functional significance for patient survival. Querying multiple gene expression databases, we show that IL13Rα2 expression increases with glioma malignancy grade, and expression for high-grade tumors is bimodal, with approximately 58% of WHO grade IV gliomas over-expressing this receptor. By several measures, IL13Rα2 expression in patient samples and low-passage primary glioma lines most consistently correlates with the expression of signature genes defining mesenchymal subclass tumors and negatively correlates with proneural signature genes as defined by two studies. Positive associations were also noted with proliferative signature genes, whereas no consistent associations were found with either classical or neural signature genes. Probing the potential functional consequences of this mesenchymal association through IPA analysis suggests that IL13Rα2 expression is associated with activation of proinflammatory and immune pathways characteristic of mesenchymal subclass tumors. In addition, survival analyses indicate that IL13Rα2 over-expression is associated with poor patient prognosis, a single gene correlation ranking IL13Rα2 in the top ~1% of total gene expression probes with regard to survival association with WHO IV gliomas. This study better defines the functional consequences of IL13Rα2 expression by demonstrating association with mesenchymal signature gene expression and poor patient prognosis. It thus highlights the utility of IL13Rα2 as a therapeutic target, and helps define patient populations most likely to respond to immunotherapy in present and future clinical trials.


Journal for ImmunoTherapy of Cancer | 2013

Chimeric antigen receptors (CARs) incorporating mutations in the IgG4 Fc spacer region to eliminate Fc receptor recognition results in improved CAR T cell persistence and anti-tumor efficacy

Mahesh Jonnalagadda; Armen Mardiros; Lauren Hoffman; Alyssa Bernanke; Wen-Chung Chang; William Bretzlaff; Renate Starr; Xiuli Wang; Julie R. Ostberg; Christine E. Brown; Stephen J. Forman

Adoptive immunotherapy using T cells genetically redirected via expression of chimeric antigen receptors (CARs) is a promising approach for cancer treatment. However, this immunotherapy is dependent in part on the optimal molecular design of the CAR, which involves an extracellular ligand-binding domain connected to an intracellular signaling domain by spacer and/or transmembrane sequences. CAR designs frequently incorporate extracellular linker regions based on the immunoglobulin constant regions of either IgG1 or IgG4. In this study we evaluated the potential for the IgG4-Fc linker to result in off-target interactions between the CAR and Fc gamma receptors (FcγRs). As proof of principle, we have focused on a CD19-specific CD19scFv-IgG4-CD28-zeta CAR, and indeed found that CAR+ T cells bound to soluble FcγRs, and did not engraft in NSG mice compared to CAR-negative T cells that only expressed an EGFRt tracking marker. We hypothesized that mutations to avoid FcγR interactions would improve CAR+ T cell persistence and anti-tumor efficacy. To this end, we generated a CD19-specific CAR that has been mutated at two sites within the CH2 region (L235E; N297Q) of the IgG4 Fc spacer, here called CD19R(EQ), as well as a CD19-specific CAR that has a CH2 deletion in its IgG4 Fc spacer (CD19Rch2Δ). These mutations/deletion do not alter the functional ability of the CAR, when expressed by T cells, to mediate antigen-specific lysis of tumor cells. However, compared to T cells that express a non-mutated CAR, T cells expressing the CD19R(EQ) and CD19Rch2Δ exhibit impaired binding to recombinant soluble FcγRs. These CD19R(EQ) and CD19Rch2Δ T cells also exhibit improved engraftment in NSG mice. Indeed the engraftment levels seen with the mutated CAR were similar to that seen with CAR-negative T cells that only expressed the EGFRt tracking marker. Importantly, elimination of CAR/FcγR interactions also significantly improves CD19-specific CAR+ T cell anti-lymphoma efficacy in NSG mice. These studies provide evidence that optimal CAR function necessitates the elimination of cellular FcγR interactions in order to improve T cell persistence and anti-tumor responses.


Journal for ImmunoTherapy of Cancer | 2015

Clinical development of IL13Rα2-targeting CAR T cells for the treatment of glioblastoma

Christine E. Brown; Renate Starr; Brenda Aguilar; Alfonso Brito; Brenda Chang; Aniee Sarkissian; Lihong Weng; Michael C. Jensen; Michael E. Barish; Behnam Badie; Stephen J. Forman

T cell immunotherapy is emerging as a powerful strategy to treat cancer, and may offer new opportunities to improve outcomes for patients with glioblastoma (GBM). Our group has developed a chimeric antigen receptor (CAR) T cell immunotherapy for GBM targeting IL-13 receptor α2 (IL13Rα2), a cell surface receptor over-expressed by the majority of high-grade gliomas. Towards this end, we have optimized IL13Rα2-specific CAR T cells by incorporating enhancements in CAR design and T cell engineering to improve T cell persistence and antitumor potency. These include a second-generation CAR containing the 41BB (CD137) costimulatory signaling domain (IL13BBζ-CAR), and a manufacturing strategy using an enriched central memory T cell (TCM) population for genetic engineering. To model clinical conditions and translate this therapy to patients we used orthotopic human GBM models with low-passage patient-derived tumor sphere (TS) lines in NSG mice. We demonstrate here that single injections of optimized IL13BBζ-CAR TCM results in significantly improved antitumor activity and T cell persistence as compared to the early-generation IL13Rα2-CAR T cells that were themselves capable of mediating transient anti-tumor activity in patients. Evaluating routes for delivery, intravenous (i.v.) versus intracranial (i.c.), we find that i.c. delivery of the therapeutic CAR T cells elicits superior antitumor efficacy as compared to i.v. administration, which in these studies provided no apparent therapeutic benefit. We also explored the capacity of T cells to traffic within the brain parenchyma, and using a multifocal disease model establish that CAR T cells injected i.c. at one tumor site are able to traffic to a second tumor site in the contralateral hemisphere. Further, we investigated variations in cell product composition, including CD4 to CD8 ratios, and observed greater tumor recurrence in mice that received 100% CD8+ CAR T cells, indicating that inclusion of CD4+ CAR T cell subsets improves the durability of the therapeutic response. Finally, we evaluated the impact of corticosteroid, given its frequent use in clinical management of GBM, and demonstrate that low dose dexamethasone does not diminish T cell antitumor activity in vivo. These findings refine both CAR T cell product and clinical parameters for optimally translating this therapy to patients, and provide the rational for our newly initiated first-in-human phase I clinical trial evaluating intracranial administration of IL13Rα2-specific CD4+ CD8+ IL13BBζ-CAR TCM for the treatment of GBM.


PLOS ONE | 2014

Cytokine induction of VCAM-1 but not IL13Rα2 on glioma cells: a tale of two antibodies.

Vaidehi Mahadev; Renate Starr; Sarah Wright; Catalina Martinez; Michael C. Jensen; Michael E. Barish; Stephen J. Forman; Christine E. Brown

The interleukin-13 receptor alpha2 (IL13Rα2) is a cell surface receptor that is over-expressed by a subset of high-grade gliomas, but not expressed at significant levels by normal brain tissue. For both malignant and non-malignant cells, IL13Rα2 surface expression is reported to be induced by various cytokines such as IL-4 or IL-13 and tumor necrosis factor (TNF). Our group has developed a therapeutic platform to target IL13Rα2-positive brain tumors by engineering human cytotoxic T lymphocytes (CTLs) to express the IL13-zetakine chimeric antigen receptor. We therefore sought to investigate the potential of cytokine stimulation to induce IL13Rα2 cell surface expression, and thereby increase susceptibility to IL13Rα2-specific T cell killing. In the course of these experiments, we unexpectedly found that the commercially available putative IL13Rα2-specific monoclonal antibody B-D13 recognizes cytokine-induced VCAM-1 on glioblastoma. We provide evidence that the induced receptor is not IL13Rα2, because its expression does not consistently correlate with IL13Rα2 mRNA levels, it does not bind IL-13, and it is not recognized by IL13-zetakine CTL. Instead we demonstrate by immunoprecipitation experiments and mass spectrometry that the antigen recognized by the B-D13 antibody following cytokine stimulation is VCAM-1, and that VCAM-1, but not IL13Rα2, is induced on glioma cells by TNF alone or in combination with IL-13 or IL-4. Further evaluation of several commercial B-D13 antibodies revealed that B-D13 is bi-specific, recognizing both IL13Rα2 and VCAM-1. This binding is non-overlapping based on soluble receptor competition experiments, and mass spectrometry identifies two distinct heavy and light chain species, providing evidence that the B-D13 reagent is di-clonal. PE-conjugation of the B-D13 antibody appears to disrupt IL13Rα2 recognition, while maintaining VCAM-1 specificity. While this work calls into question previous studies that have used the B-D13 antibody to assess IL13Rα2 expression, it also suggests that TNF may have significant effects on glioma biology by up-regulating VCAM-1.

Collaboration


Dive into the Renate Starr's collaboration.

Top Co-Authors

Avatar

Christine E. Brown

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Stephen J. Forman

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Brenda Aguilar

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Behnam Badie

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Michael C. Jensen

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Michael E. Barish

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Aniee Sarkissian

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Wen-Chung Chang

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Alfonso Brito

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Brenda Chang

City of Hope National Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge