Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
N Engl J Med ; 375(26): 2561-9, 2016 12 29.
Article in English | MEDLINE | ID: mdl-28029927

ABSTRACT

A patient with recurrent multifocal glioblastoma received chimeric antigen receptor (CAR)-engineered T cells targeting the tumor-associated antigen interleukin-13 receptor alpha 2 (IL13Rα2). Multiple infusions of CAR T cells were administered over 220 days through two intracranial delivery routes - infusions into the resected tumor cavity followed by infusions into the ventricular system. Intracranial infusions of IL13Rα2-targeted CAR T cells were not associated with any toxic effects of grade 3 or higher. After CAR T-cell treatment, regression of all intracranial and spinal tumors was observed, along with corresponding increases in levels of cytokines and immune cells in the cerebrospinal fluid. This clinical response continued for 7.5 months after the initiation of CAR T-cell therapy. (Funded by Gateway for Cancer Research and others; ClinicalTrials.gov number, NCT02208362 .).


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Glioblastoma/therapy , Immunotherapy, Adoptive , Neoplasm Recurrence, Local/therapy , Receptors, Antigen, T-Cell/therapeutic use , Cell Engineering , Combined Modality Therapy , Humans , Interleukin-13 Receptor alpha2 Subunit , Male , Middle Aged
2.
Mol Ther ; 26(1): 31-44, 2018 01 03.
Article in English | MEDLINE | ID: mdl-29103912

ABSTRACT

T cell immunotherapy is emerging as a powerful strategy to treat cancer and may improve outcomes for patients with glioblastoma (GBM). We have developed a chimeric antigen receptor (CAR) T cell immunotherapy targeting IL-13 receptor α2 (IL13Rα2) for the treatment of GBM. Here, we describe the optimization of IL13Rα2-targeted CAR T cells, including the design of a 4-1BB (CD137) co-stimulatory CAR (IL13BBζ) and a manufacturing platform using enriched central memory T cells. Utilizing orthotopic human GBM models with patient-derived tumor sphere lines in NSG mice, we found that IL13BBζ-CAR T cells improved anti-tumor activity and T cell persistence as compared to first-generation IL13ζ-CAR CD8+ T cells that had shown evidence for bioactivity in patients. Investigating the impact of corticosteroids, given their frequent use in the clinical management of GBM, we demonstrate that low-dose dexamethasone does not diminish CAR T cell anti-tumor activity in vivo. Furthermore, we found that local intracranial delivery of CAR T cells elicits superior anti-tumor efficacy as compared to intravenous administration, with intraventricular infusions exhibiting possible benefit over intracranial tumor infusions in a multifocal disease model. Overall, these findings help define parameters for the clinical translation of CAR T cell therapy for the treatment of brain tumors.


Subject(s)
Glioblastoma/immunology , Glioblastoma/metabolism , Immunotherapy, Adoptive , Interleukin-13 Receptor alpha2 Subunit/antagonists & inhibitors , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Antibodies, Neoplasm/immunology , Antigens, CD19/immunology , Brain Neoplasms/immunology , Brain Neoplasms/metabolism , Brain Neoplasms/therapy , Cytotoxicity, Immunologic , Dextroamphetamine/pharmacology , Disease Models, Animal , Gene Order , Genetic Engineering , Genetic Vectors/genetics , Glioblastoma/mortality , Glioblastoma/therapy , Humans , Immunotherapy, Adoptive/methods , Interleukin-13 Receptor alpha2 Subunit/immunology , Mice , Receptors, Chimeric Antigen/chemistry , Xenograft Model Antitumor Assays
3.
Mol Ther ; 23(4): 757-68, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25366031

ABSTRACT

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.


Subject(s)
Immunoglobulin G/immunology , Mutant Chimeric Proteins/metabolism , Mutation , Neoplasms, Experimental/therapy , Receptors, Antigen/metabolism , Receptors, Fc/metabolism , T-Lymphocytes/immunology , Animals , Immunotherapy , Mice , Neoplasms, Experimental/immunology , Protein Binding , Receptors, Antigen/genetics
4.
Blood ; 122(18): 3138-48, 2013 Oct 31.
Article in English | MEDLINE | ID: mdl-24030378

ABSTRACT

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.


Subject(s)
Cytotoxicity, Immunologic/immunology , Interleukin-3 Receptor alpha Subunit/immunology , Leukemia, Myeloid/immunology , Receptors, Antigen/immunology , Single-Chain Antibodies/immunology , T-Lymphocytes/immunology , Acute Disease , Animals , Cell Line , Cell Line, Tumor , Coculture Techniques , Cytokines/immunology , Cytokines/metabolism , Flow Cytometry , HEK293 Cells , Humans , Immunotherapy, Adoptive/methods , Interleukin-3 Receptor alpha Subunit/genetics , Interleukin-3 Receptor alpha Subunit/metabolism , K562 Cells , Leukemia, Myeloid/pathology , Leukemia, Myeloid/therapy , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Receptors, Antigen/genetics , Receptors, Antigen/metabolism , Single-Chain Antibodies/genetics , Single-Chain Antibodies/metabolism , T-Lymphocytes/metabolism , T-Lymphocytes/transplantation , Xenograft Model Antitumor Assays
5.
Nat Med ; 30(4): 1001-1012, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38454126

ABSTRACT

Chimeric antigen receptor T cell (CAR-T) therapy is an emerging strategy to improve treatment outcomes for recurrent high-grade glioma, a cancer that responds poorly to current therapies. Here we report a completed phase I trial evaluating IL-13Rα2-targeted CAR-T cells in 65 patients with recurrent high-grade glioma, the majority being recurrent glioblastoma (rGBM). Primary objectives were safety and feasibility, maximum tolerated dose/maximum feasible dose and a recommended phase 2 dose plan. Secondary objectives included overall survival, disease response, cytokine dynamics and tumor immune contexture biomarkers. This trial evolved to evaluate three routes of locoregional T cell administration (intratumoral (ICT), intraventricular (ICV) and dual ICT/ICV) and two manufacturing platforms, culminating in arm 5, which utilized dual ICT/ICV delivery and an optimized manufacturing process. Locoregional CAR-T cell administration was feasible and well tolerated, and as there were no dose-limiting toxicities across all arms, a maximum tolerated dose was not determined. Probable treatment-related grade 3+ toxicities were one grade 3 encephalopathy and one grade 3 ataxia. A clinical maximum feasible dose of 200 × 106 CAR-T cells per infusion cycle was achieved for arm 5; however, other arms either did not test or achieve this dose due to manufacturing feasibility. A recommended phase 2 dose will be refined in future studies based on data from this trial. Stable disease or better was achieved in 50% (29/58) of patients, with two partial responses, one complete response and a second complete response after additional CAR-T cycles off protocol. For rGBM, median overall survival for all patients was 7.7 months and for arm 5 was 10.2 months. Central nervous system increases in inflammatory cytokines, including IFNγ, CXCL9 and CXCL10, were associated with CAR-T cell administration and bioactivity. Pretreatment intratumoral CD3 T cell levels were positively associated with survival. These findings demonstrate that locoregional IL-13Rα2-targeted CAR-T therapy is safe with promising clinical activity in a subset of patients. ClinicalTrials.gov Identifier: NCT02208362 .


Subject(s)
Glioblastoma , Glioma , Receptors, Chimeric Antigen , Humans , Neoplasm Recurrence, Local , Glioma/therapy , T-Lymphocytes , Glioblastoma/therapy , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods
6.
Blood ; 118(5): 1255-63, 2011 Aug 04.
Article in English | MEDLINE | ID: mdl-21653320

ABSTRACT

An unmet need in cell engineering is the availability of a single transgene encoded, functionally inert, human polypeptide that can serve multiple purposes, including ex vivo cell selection, in vivo cell tracking, and as a target for in vivo cell ablation. Here we describe a truncated human EGFR polypeptide (huEGFRt) that is devoid of extracellular N-terminal ligand binding domains and intracellular receptor tyrosine kinase activity but retains the native amino acid sequence, type I transmembrane cell surface localization, and a conformationally intact binding epitope for pharmaceutical-grade anti-EGFR monoclonal antibody, cetuximab (Erbitux). After lentiviral transduction of human T cells with vectors that coordinately express tumor-specific chimeric antigen receptors and huEGFRt, we show that huEGFRt serves as a highly efficient selection epitope for chimeric antigen receptor(+) T cells using biotinylated cetuximab in conjunction with current good manufacturing practices (cGMP)-grade anti-biotin immunomagnetic microbeads. Moreover, huEGFRt provides a cell surface marker for in vivo tracking of adoptively transferred T cells using both flow cytometry and immunohistochemistry, and a target for cetuximab-mediated antibody-dependent cellular cytotoxicity and in vivo elimination. The versatility of huEGFRt and the availability of pharmaceutical-grade reagents for its clinical application denote huEGFRt as a significant new tool for cellular engineering.


Subject(s)
Antigens, Surface/genetics , Apoptosis/genetics , Cell Separation/methods , Cell Tracking/methods , Tissue Engineering/methods , Transgenes/genetics , Animals , Cells/metabolism , Cells/pathology , Cells, Cultured , Genes, Reporter , Genes, Transgenic, Suicide/physiology , Genes, erbB-1 , Humans , Interleukin Receptor Common gamma Subunit/genetics , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Peptides/genetics
7.
Cancer Res Commun ; 3(1): 66-79, 2023 01.
Article in English | MEDLINE | ID: mdl-36968221

ABSTRACT

Chimeric antigen receptor (CAR) T cell immunotherapy is emerging as a powerful strategy for cancer therapy; however, an important safety consideration is the potential for off-tumor recognition of normal tissue. This is particularly important as ligand-based CARs are optimized for clinical translation. Our group has developed and clinically translated an IL13(E12Y) ligand-based CAR targeting the cancer antigen IL13Rα2 for treatment of glioblastoma (GBM). There remains limited understanding of how IL13-ligand CAR design impacts the activity and selectivity for the intended tumor-associated target IL13Rα2 versus the more ubiquitous unintended target IL13Rα1. In this study, we functionally compared IL13(E12Y)-CARs incorporating different intracellular signaling domains, including first-generation CD3ζ-containing CARs (IL13ζ), second-generation 4-1BB (CD137)-containing or CD28-containing CARs (IL13-BBζ or IL13-28ζ), and third-generation CARs containing both 4-1BB and CD28 (IL13-28BBζ). In vitro coculture assays at high tumor burden establish that second-generation IL13-BBζ or IL13-28ζ outperform first-generation IL13ζ and third-generation IL13-28BBζ CAR designs, with IL13-BBζ providing superior CAR proliferation and in vivo antitumor potency in human xenograft mouse models. IL13-28ζ displayed a lower threshold for antigen recognition, resulting in higher off-target IL13Rα1 reactivity both in vitro and in vivo. Syngeneic mouse models of GBM also demonstrate safety and antitumor potency of murine IL13-BBζ CAR T cells delivered systemically after lymphodepletion. These findings support the use of IL13-BBζ CARs for greater selective recognition of IL13Rα2 over IL13Rα1, higher proliferative potential, and superior antitumor responsiveness. This study exemplifies the potential of modulating factors outside the antigen targeting domain of a CAR to improve selective tumor recognition. Significance: This study reveals how modulating CAR design outside the antigen targeting domain improves selective tumor recognition. Specifically, this work shows improved specificity, persistence, and efficacy of 4-1BB-based IL13-ligand CARs. Human clinical trials evaluating IL13-41BB-CAR T cells are ongoing, supporting the clinical significance of these findings.


Subject(s)
Glioblastoma , Interleukin-13 Receptor alpha2 Subunit , Receptors, Chimeric Antigen , Humans , Mice , Animals , Receptors, Antigen, T-Cell , Receptors, Chimeric Antigen/genetics , T-Lymphocytes , Immunotherapy, Adoptive/methods , Interleukin-13 Receptor alpha2 Subunit/genetics , Interleukin-13/genetics , CD28 Antigens/genetics , Ligands , Glioblastoma/therapy , Disease Models, Animal
8.
Cell Stem Cell ; 29(4): 515-527.e8, 2022 04 07.
Article in English | MEDLINE | ID: mdl-35278370

ABSTRACT

Unlimited generation of chimeric antigen receptor (CAR) T cells from human-induced pluripotent stem cells (iPSCs) is an attractive approach for "off-the-shelf" CAR T cell immunotherapy. Approaches to efficiently differentiate iPSCs into canonical αß T cell lineages, while maintaining CAR expression and functionality, however, have been challenging. We report that iPSCs reprogramed from CD62L+ naive and memory T cells followed by CD19-CAR engineering and 3D-organoid system differentiation confers products with conventional CD8αß-positive CAR T cell characteristics. Expanded iPSC CD19-CAR T cells showed comparable antigen-specific activation, degranulation, cytotoxicity, and cytokine secretion compared with conventional CD19-CAR T cells and maintained homogeneous expression of the TCR derived from the initial clone. iPSC CD19-CAR T cells also mediated potent antitumor activity in vivo, prolonging survival of mice with CD19+ human tumor xenografts. Our study establishes feasible methodologies to generate highly functional CAR T cells from iPSCs to support the development of "off-the-shelf" manufacturing strategies.


Subject(s)
Induced Pluripotent Stem Cells , Receptors, Chimeric Antigen , Animals , Cell Differentiation , Humans , Immunotherapy , Immunotherapy, Adoptive/methods , Induced Pluripotent Stem Cells/metabolism , Mice , Organoids/metabolism , Receptors, Chimeric Antigen/metabolism
9.
Neuro Oncol ; 24(8): 1318-1330, 2022 08 01.
Article in English | MEDLINE | ID: mdl-35100373

ABSTRACT

BACKGROUND: Wide-spread application of chimeric antigen receptor (CAR) T cell therapy for cancer is limited by the current use of autologous CAR T cells necessitating the manufacture of individualized therapeutic products for each patient. To address this challenge, we have generated an off-the-shelf, allogeneic CAR T cell product for the treatment of glioblastoma (GBM), and present here the feasibility, safety, and therapeutic potential of this approach. METHODS: We generated for clinical use a healthy-donor derived IL13Rα2-targeted CAR+ (IL13-zetakine+) cytolytic T-lymphocyte (CTL) product genetically engineered using zinc finger nucleases (ZFNs) to permanently disrupt the glucocorticoid receptor (GR) (GRm13Z40-2) and endow resistance to glucocorticoid treatment. In a phase I safety and feasibility trial we evaluated these allogeneic GRm13Z40-2 T cells in combination with intracranial administration of recombinant human IL-2 (rhIL-2; aldesleukin) in six patients with unresectable recurrent GBM that were maintained on systemic dexamethasone (4-12 mg/day). RESULTS: The GRm13Z40-2 product displayed dexamethasone-resistant effector activity without evidence for in vitro alloreactivity. Intracranial administration of GRm13Z40-2 in four doses of 108 cells over a two-week period with aldesleukin (9 infusions ranging from 2500-5000 IU) was well tolerated, with indications of transient tumor reduction and/or tumor necrosis at the site of T cell infusion in four of the six treated research subjects. Antibody reactivity against GRm13Z40-2 cells was detected in the serum of only one of the four tested subjects. CONCLUSIONS: This first-in-human experience establishes a foundation for future adoptive therapy studies using off-the-shelf, zinc-finger modified, and/or glucocorticoid resistant CAR T cells.


Subject(s)
Glioblastoma , Interleukin-13 Receptor alpha2 Subunit , Dexamethasone , Glioblastoma/pathology , Glucocorticoids , Humans , Immunotherapy, Adoptive , Steroids , T-Lymphocytes , Xenograft Model Antitumor Assays
10.
Biol Blood Marrow Transplant ; 16(9): 1245-56, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20304086

ABSTRACT

Immunotherapeutic ablation of lymphoma is a conceptually attractive treatment strategy that is the subject of intense translational research. Cytotoxic T lymphocytes (CTLs) that are genetically modified to express CD19- or CD20-specific, single-chain antibody-derived chimeric antigen receptors (CARs) display HLA-independent antigen-specific recognition/killing of lymphoma targets. Here, we describe our initial experience in applying CAR-redirected autologous CTL adoptive therapy to patients with recurrent lymphoma. Using plasmid vector electrotransfer/drug selection systems, cloned and polyclonal CAR(+) CTLs were generated from autologous peripheral blood mononuclear cells and expanded in vitro to cell numbers sufficient for clinical use. In 2 FDA-authorized trials, patients with recurrent diffuse large cell lymphoma were treated with cloned CD8(+) CTLs expressing a CD20-specific CAR (along with NeoR) after autologous hematopoietic stem cell transplantation, and patients with refractory follicular lymphoma were treated with polyclonal T cell preparations expressing a CD19-specific CAR (along with HyTK, a fusion of hygromycin resistance and HSV-1 thymidine kinase suicide genes) and low-dose s.c. recombinant human interleukin-2. A total of 15 infusions were administered (5 at 10(8)cells/m(2), 7 at 10(9)cells/m(2), and 3 at 2 x 10(9)cells/m(2)) to 4 patients. Overt toxicities attributable to CTL administration were not observed; however, detection of transferred CTLs in the circulation, as measured by quantitative polymerase chain reaction, was short (24 hours to 7 days), and cellular antitransgene immune rejection responses were noted in 2 patients. These studies reveal the primary barrier to therapeutic efficacy is limited persistence, and provide the rationale to prospectively define T cell populations intrinsically programmed for survival after adoptive transfer and to modulate the immune status of recipients to prevent/delay antitransgene rejection responses.


Subject(s)
Adoptive Transfer/methods , Antigens, CD19/immunology , Antigens, CD20/immunology , Lymphoma, B-Cell/therapy , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/transplantation , Transgenes/immunology , Antigens, CD19/biosynthesis , Antigens, CD19/genetics , Antigens, CD20/biosynthesis , Antigens, CD20/genetics , Humans , Immune Tolerance , Lymphoma, B-Cell/immunology , Lymphoma, Follicular/immunology , Lymphoma, Follicular/therapy , Lymphoma, Large B-Cell, Diffuse/immunology , Lymphoma, Large B-Cell, Diffuse/therapy , Receptors, Antigen, T-Cell/biosynthesis , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes, Cytotoxic/physiology , Transfection
11.
Sci Transl Med ; 12(533)2020 03 04.
Article in English | MEDLINE | ID: mdl-32132216

ABSTRACT

Although chimeric antigen receptor (CAR) T cells have demonstrated signs of antitumor activity against glioblastoma (GBM), tumor heterogeneity remains a critical challenge. To achieve broader and more effective GBM targeting, we developed a peptide-bearing CAR exploiting the GBM-binding potential of chlorotoxin (CLTX). We find that CLTX peptide binds a great proportion of tumors and constituent tumor cells. CAR T cells using CLTX as the targeting domain (CLTX-CAR T cells) mediate potent anti-GBM activity and efficiently target tumors lacking expression of other GBM-associated antigens. Treatment with CLTX-CAR T cells resulted in tumor regression in orthotopic xenograft GBM tumor models. CLTX-CAR T cells do not exhibit observable off-target effector activity against normal cells or after adoptive transfer into mice. Effective targeting by CLTX-CAR T cells requires cell surface expression of matrix metalloproteinase-2. Our results pioneer a peptide toxin in CAR design, expanding the repertoire of tumor-selective CAR T cells with the potential to reduce antigen escape.


Subject(s)
Glioblastoma , Scorpion Venoms , Animals , Cell Line, Tumor , Glioblastoma/therapy , Immunotherapy, Adoptive , Matrix Metalloproteinase 2 , Mice , Receptors, Antigen, T-Cell , T-Lymphocytes , Xenograft Model Antitumor Assays
13.
J Leukoc Biol ; 82(5): 1322-31, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17711975

ABSTRACT

Circulating NK cells normally experience temperature gradients as they move about the body, but the onset of inflammation can expose them and their targets to febrile temperatures for several hours. We found that exposure of human peripheral blood NK cells and target cells to fever-range temperatures significantly enhances lysis of Colo205 target cells. A similar effect was not observed when NK cell lines or IL-2-activated peripheral blood NK cells were used as effectors, indicating that thermal sensitivity of effectors is maturation or activation state-dependent. Use of blocking antibodies revealed that this effect is also dependent on the function of the activating receptor NKG2D and its ligand MHC class I-related chain A (MICA). On NK cells, it was observed that thermal exposure does not affect the total level of NKG2D surface expression, but does result in its distinct clustering, identical to that which occurs following IL-2-induced activation. On tumor target cells, a similar, mild temperature elevation results in transcriptional up-regulation of MICA in a manner that correlates with increased sensitivity to cytolysis. Overall, these data reveal that NK cells possess thermally responsive regulatory elements, which facilitate their ability to capitalize on reciprocal, stress-induced changes simultaneously occurring on target cells during inflammation and fever.


Subject(s)
Cell Membrane/metabolism , Cell Survival/immunology , Cytotoxicity, Immunologic , Histocompatibility Antigens Class I/genetics , Hot Temperature , Killer Cells, Natural/cytology , Receptors, Immunologic/metabolism , Cell Communication , Cell Survival/physiology , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Fluorescent Antibody Technique , Histocompatibility Antigens Class I/metabolism , Humans , Immunologic Surveillance , Interleukin-2/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , NK Cell Lectin-Like Receptor Subfamily K , Neoplasms/metabolism , Neoplasms/pathology , Protein Transport , Receptors, Immunologic/genetics , Receptors, Natural Killer Cell , Regulatory Elements, Transcriptional , Reverse Transcriptase Polymerase Chain Reaction
14.
JCI Insight ; 3(10)2018 05 17.
Article in English | MEDLINE | ID: mdl-29769444

ABSTRACT

Chimeric antigen receptor-modified (CAR-modified) T cells have shown promising therapeutic effects for hematological malignancies, yet limited and inconsistent efficacy against solid tumors. The refinement of CAR therapy requires an understanding of the optimal characteristics of the cellular products, including the appropriate composition of CD4+ and CD8+ subsets. Here, we investigated the differential antitumor effect of CD4+ and CD8+ CAR T cells targeting glioblastoma-associated (GBM-associated) antigen IL-13 receptor α2 (IL13Rα2). Upon stimulation with IL13Rα2+ GBM cells, the CD8+ CAR T cells exhibited robust short-term effector function but became rapidly exhausted. By comparison, the CD4+ CAR T cells persisted after tumor challenge and sustained their effector potency. Mixing with CD4+ CAR T cells failed to ameliorate the effector dysfunction of CD8+ CAR T cells, while surprisingly, CD4+ CAR T cell effector potency was impaired when coapplied with CD8+ T cells. In orthotopic GBM models, CD4+ outperformed CD8+ CAR T cells, especially for long-term antitumor response. Further, maintenance of the CD4+ subset was positively correlated with the recursive killing ability of CAR T cell products derived from GBM patients. These findings identify CD4+ CAR T cells as a highly potent and clinically important T cell subset for effective CAR therapy.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Glioblastoma/pathology , Humans
15.
Clin Cancer Res ; 24(1): 95-105, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29061641

ABSTRACT

Purpose: Metastasis to the brain from breast cancer remains a significant clinical challenge, and may be targeted with CAR-based immunotherapy. CAR design optimization for solid tumors is crucial due to the absence of truly restricted antigen expression and potential safety concerns with "on-target off-tumor" activity. Here, we have optimized HER2-CAR T cells for the treatment of breast to brain metastases, and determined optimal second-generation CAR design and route of administration for xenograft mouse models of breast metastatic brain tumors, including multifocal and leptomeningeal disease.Experimental Design: HER2-CAR constructs containing either CD28 or 4-1BB intracellular costimulatory signaling domains were compared for functional activity in vitro by measuring cytokine production, T-cell proliferation, and tumor killing capacity. We also evaluated HER2-CAR T cells delivered by intravenous, local intratumoral, or regional intraventricular routes of administration using in vivo human xenograft models of breast cancer that have metastasized to the brain.Results: Here, we have shown that HER2-CARs containing the 4-1BB costimulatory domain confer improved tumor targeting with reduced T-cell exhaustion phenotype and enhanced proliferative capacity compared with HER2-CARs containing the CD28 costimulatory domain. Local intracranial delivery of HER2-CARs showed potent in vivo antitumor activity in orthotopic xenograft models. Importantly, we demonstrated robust antitumor efficacy following regional intraventricular delivery of HER2-CAR T cells for the treatment of multifocal brain metastases and leptomeningeal disease.Conclusions: Our study shows the importance of CAR design in defining an optimized CAR T cell, and highlights intraventricular delivery of HER2-CAR T cells for treating multifocal brain metastases. Clin Cancer Res; 24(1); 95-105. ©2017 AACR.


Subject(s)
Brain Neoplasms/immunology , Brain Neoplasms/secondary , Breast Neoplasms/pathology , Immunotherapy, Adoptive , Receptors, Antigen, T-Cell/metabolism , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Brain Neoplasms/therapy , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Breast Neoplasms/metabolism , CD28 Antigens/genetics , Cytokines/metabolism , Cytotoxicity, Immunologic , Disease Models, Animal , Female , Humans , Immunotherapy, Adoptive/methods , Inflammation Mediators/metabolism , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Receptor, ErbB-2/genetics , Receptors, Chimeric Antigen/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Tumor Necrosis Factor Receptor Superfamily, Member 9/genetics , Xenograft Model Antitumor Assays
18.
PLoS One ; 11(1): e0146885, 2016.
Article in English | MEDLINE | ID: mdl-26761817

ABSTRACT

New therapeutic modalities are needed for ovarian cancer, the most lethal gynecologic malignancy. Recent clinical trials have demonstrated the impressive therapeutic potential of adoptive therapy using chimeric antigen receptor (CAR)-redirected T cells to target hematological cancers, and emerging studies suggest a similar impact may be achieved for solid cancers. We sought determine whether genetically-modified T cells targeting the CE7-epitope of L1-CAM, a cell adhesion molecule aberrantly expressed in several cancers, have promise as an immunotherapy for ovarian cancer, first demonstrating that L1-CAM was highly over-expressed on a panel of ovarian cancer cell lines, primary ovarian tumor tissue specimens, and ascites-derived primary cancer cells. Human central memory derived T cells (TCM) were then genetically modified to express an anti-L1-CAM CAR (CE7R), which directed effector function upon tumor antigen stimulation as assessed by in vitro cytokine secretion and cytotoxicity assays. We also found that CE7R+ T cells were able to target primary ovarian cancer cells. Intraperitoneal (i.p.) administration of CE7R+ TCM induced a significant regression of i.p. established SK-OV-3 xenograft tumors in mice, inhibited ascites formation, and conferred a significant survival advantage compared with control-treated animals. Taken together, these studies indicate that adoptive transfer of L1-CAM-specific CE7R+ T cells may offer a novel and effective immunotherapy strategy for advanced ovarian cancer.


Subject(s)
Gene Expression Regulation, Neoplastic , Neural Cell Adhesion Molecule L1/metabolism , Ovarian Neoplasms/metabolism , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Animals , Cell Adhesion , Cell Line, Tumor , Epitopes, T-Lymphocyte/immunology , Female , Humans , Immunohistochemistry , Immunotherapy, Adoptive , In Situ Hybridization, Fluorescence , Lentivirus/genetics , Leukocyte Common Antigens/metabolism , Mice , Mice, Inbred NOD , Microscopy, Confocal , Neoplasm Transplantation
19.
Cell Cycle ; 14(14): 2340-54, 2015.
Article in English | MEDLINE | ID: mdl-26131730

ABSTRACT

Maintenance of normal core body temperature is vigorously defended by long conserved, neurovascular homeostatic mechanisms that assist in heat dissipation during prolonged, heat generating exercise or exposure to warm environments. Moreover, during febrile episodes, body temperature can be significantly elevated for at least several hours at a time. Thus, as blood cells circulate throughout the body, physiologically relevant variations in surrounding tissue temperature can occur; moreover, shifts in core temperature occur during daily circadian cycles. This study has addressed the fundamental question of whether the threshold of stimulation needed to activate lymphocytes is influenced by temperature increases associated with physiologically relevant increases in temperature. We report that the need for co-stimulation of CD4+ T cells via CD28 ligation for the production of IL-2 is significantly reduced when cells are exposed to fever-range temperature. Moreover, even in the presence of sufficient CD28 ligation, provision of extra heat further increases IL-2 production. Additional in vivo and in vitro data (using both thermal and chemical modulation of membrane fluidity) support the hypothesis that the mechanism by which temperature modulates co-stimulation is linked to increases in membrane fluidity and membrane macromolecular clustering in the plasma membrane. Thermally-regulated changes in plasma membrane organization in response to physiological increases in temperature may assist in the geographical control of lymphocyte activation, i.e., stimulating activation in lymph nodes rather than in cooler surface regions, and further, may temporarily and reversibly enable CD4+ T cells to become more quickly and easily activated during times of infection during fever.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Animals , Ankyrins/metabolism , CD28 Antigens/deficiency , CD28 Antigens/genetics , CD28 Antigens/metabolism , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Enzyme-Linked Immunosorbent Assay , Fluorescence Polarization , Humans , Interleukin-2/analysis , Interleukin-2/genetics , Jurkat Cells , Lymphocyte Activation/drug effects , Membrane Fluidity/drug effects , Mice , Mice, Inbred BALB C , Mice, Knockout , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Spectrin/metabolism , Temperature , Tetradecanoylphorbol Acetate/pharmacology , beta-Cyclodextrins/pharmacology
20.
Clin Cancer Res ; 21(18): 4062-72, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26059190

ABSTRACT

PURPOSE: A first-in-human pilot safety and feasibility trial evaluating chimeric antigen receptor (CAR)-engineered, autologous primary human CD8(+) cytotoxic T lymphocytes (CTL) targeting IL13Rα2 for the treatment of recurrent glioblastoma (GBM). EXPERIMENTAL DESIGN: Three patients with recurrent GBM were treated with IL13(E13Y)-zetakine CD8(+) CTL targeting IL13Rα2. Patients received up to 12 local infusions at a maximum dose of 10(8) CAR-engineered T cells via a catheter/reservoir system. RESULTS: We demonstrate the feasibility of manufacturing sufficient numbers of autologous CTL clones expressing an IL13(E13Y)-zetakine CAR for redirected HLA-independent IL13Rα2-specific effector function for a cohort of patients diagnosed with GBM. Intracranial delivery of the IL13-zetakine(+) CTL clones into the resection cavity of 3 patients with recurrent disease was well-tolerated, with manageable temporary brain inflammation. Following infusion of IL13-zetakine(+) CTLs, evidence for transient anti-glioma responses was observed in 2 of the patients. Analysis of tumor tissue from 1 patient before and after T-cell therapy suggested reduced overall IL13Rα2 expression within the tumor following treatment. MRI analysis of another patient indicated an increase in tumor necrotic volume at the site of IL13-zetakine(+) T-cell administration. CONCLUSIONS: These findings provide promising first-in-human clinical experience for intracranial administration of IL13Rα2-specific CAR T cells for the treatment of GBM, establishing a foundation on which future refinements of adoptive CAR T-cell therapies can be applied.


Subject(s)
Brain Neoplasms/therapy , CD8-Positive T-Lymphocytes/immunology , Glioblastoma/therapy , Immunotherapy, Adoptive/methods , Interleukin-13 Receptor alpha2 Subunit/therapeutic use , Receptors, Antigen, T-Cell/therapeutic use , Adult , Aged , Brain/pathology , Brain Neoplasms/immunology , CD8-Positive T-Lymphocytes/cytology , Feasibility Studies , Female , Glioblastoma/immunology , Glioma/immunology , Glioma/therapy , HLA Antigens/chemistry , Humans , Inflammation , Magnetic Resonance Imaging , Male , Middle Aged , Patient Safety , Pilot Projects , Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell/immunology , Recurrence , Treatment Outcome , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL