Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters











Publication year range
1.
World Neurosurg ; 125: e487-e496, 2019 05.
Article in English | MEDLINE | ID: mdl-30710723

ABSTRACT

BACKGROUND: Few prognostic markers are available for patients with non-small-cell lung cancer (NSCLC) undergoing neurosurgical resection of symptomatic brain metastases. OBJECTIVE: We investigated whether tumor mutation status (EGFR, KRAS, ALK, ROS1, and BRAF) and treatment history were associated with survival after neurosurgery. METHODS: We reviewed the electronic health records of 104 patients with NSCLC with genomic profiling who underwent neurosurgical resection for symptomatic brain metastases at an academic institution between January 2000 and January 2018. We used multivariate Cox proportional hazards regression models to evaluate the association between overall survival (OS) after neurosurgery and clinicopathologic factors, including mutation status. RESULTS: Mean age of patients in this study was 61 (±12) years, and 44% were men. The median OS after neurosurgery was 24 months (95% confidence interval, 18-34 months). Our multivariate analysis showed that the presence of an EGFR mutation in the tumor was significantly associated with improved OS (hazard ratio [HR], 0.214; P = 0.029), independent of tyrosine kinase inhibitor use. Presence of KRAS, ALK, ROS1, and BRAF alterations was not associated with survival (all P > 0.05). Conversely, older age (HR, 1.039; P = 0.029), a history of multiple brain irradiation procedures (HR, 9.197; P < 0.001), and presence of extracranial metastasis (HR, 2.556; P = 0.016) resulted in increased risk of mortality. CONCLUSIONS: Patients requiring surgical resection of an epidermal growth factor receptor-mutated NSCLC brain metastasis had an associated improved survival compared with patients without this mutation, independent of tyrosine kinase inhibitor use. Decreased survival was associated with older age, multiple previous brain radiation therapies, and extracranial metastasis.


Subject(s)
Brain Neoplasms/therapy , Carcinoma, Non-Small-Cell Lung/genetics , ErbB Receptors/genetics , Mutation/genetics , Adult , Age Factors , Aged , Aged, 80 and over , Brain Neoplasms/genetics , Brain Neoplasms/secondary , Carcinoma, Non-Small-Cell Lung/therapy , Disease-Free Survival , Female , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Male , Middle Aged , Neoplasm Metastasis , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Treatment Outcome
5.
J Clin Invest ; 126(8): 3036-52, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27427982

ABSTRACT

In preclinical models of glioblastoma, antigen escape variants can lead to tumor recurrence after treatment with CAR T cells that are redirected to single tumor antigens. Given the heterogeneous expression of antigens on glioblastomas, we hypothesized that a bispecific CAR molecule would mitigate antigen escape and improve the antitumor activity of T cells. Here, we created a CAR that joins a HER2-binding scFv and an IL13Rα2-binding IL-13 mutein to make a tandem CAR exodomain (TanCAR) and a CD28.ζ endodomain. We determined that patient TanCAR T cells showed distinct binding to HER2 or IL13Rα2 and had the capability to lyse autologous glioblastoma. TanCAR T cells exhibited activation dynamics that were comparable to those of single CAR T cells upon encounter of HER2 or IL13Rα2. We observed that TanCARs engaged HER2 and IL13Rα2 simultaneously by inducing HER2-IL13Rα2 heterodimers, which promoted superadditive T cell activation when both antigens were encountered concurrently. TanCAR T cell activity was more sustained but not more exhaustible than that of T cells that coexpressed a HER2 CAR and an IL13Rα2 CAR, T cells with a unispecific CAR, or a pooled product. In a murine glioblastoma model, TanCAR T cells mitigated antigen escape, displayed enhanced antitumor efficacy, and improved animal survival. Thus, TanCAR T cells show therapeutic potential to improve glioblastoma control by coengaging HER2 and IL13Rα2 in an augmented, bivalent immune synapse that enhances T cell functionality and reduces antigen escape.


Subject(s)
Brain Neoplasms/metabolism , Glioblastoma/metabolism , Interleukin-13 Receptor alpha2 Subunit/metabolism , Receptor, ErbB-2/metabolism , T-Lymphocytes/metabolism , Tumor Escape , Animals , Antigens, Neoplasm/metabolism , Antineoplastic Agents/chemistry , Cell Line, Tumor , Humans , Immunotherapy, Adoptive , Interleukin-13/metabolism , Lymphocyte Activation , Mice , Mice, SCID , Neoplasm Recurrence, Local , Neoplasm Transplantation , Protein Binding , Protein Multimerization , Receptors, Antigen, T-Cell/metabolism , Transgenes
6.
J Neurooncol ; 123(3): 459-64, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25877468

ABSTRACT

Glioblastoma is a devastating cancer with universally poor outcomes in spite of current standard multimodal therapy. Immunotherapy is an attractive new treatment modality given its potential for exquisite specificity and its favorable side effect profile; however, clinical trials of immunotherapy in GBM have thus far shown modest benefit. Optimally combining radiation with immunotherapy may be the key to unlocking the potential of both therapies given the evidence that radiation can enhance anti-tumor immunity. Here we review this evidence and discuss considerations for combined therapy.


Subject(s)
Brain Neoplasms/therapy , Glioblastoma/therapy , Immunotherapy/methods , Radiotherapy/methods , Brain Neoplasms/immunology , Combined Modality Therapy , Glioblastoma/immunology , Humans
7.
J Immunol Res ; 2014: 796856, 2014.
Article in English | MEDLINE | ID: mdl-24804271

ABSTRACT

Glioblastoma (GBM) is the most common primary brain tumor, and despite aggressive therapy with surgery, radiation, and chemotherapy, average survival remains at about 1.5 years. The highly infiltrative and invasive nature of GBM requires that alternative treatments for this disease be widespread and targeted to tumor cells. Immunotherapy in the form of tumor vaccines has the potential to meet this need. Vaccines against GBM hold the promise of triggering specific and systemic antitumor immune responses that may be the key to eradicating this unrelenting cancer. In this review, we will discuss past and present clinical trials of various GBM vaccines and their potential impact on the future care of GBM patients. There have been many promising phase I and phase II GBM vaccine studies that have led to ongoing and upcoming phase III trials. If the results of these randomized trials show a survival benefit, immunotherapy will become a standard part of the treatment of this devastating disease.


Subject(s)
Antigens, Neoplasm/immunology , Brain Neoplasms/prevention & control , Cancer Vaccines/immunology , Clinical Trials as Topic , Glioblastoma/prevention & control , Adjuvants, Immunologic/administration & dosage , Antibodies, Monoclonal/therapeutic use , Brain Neoplasms/immunology , Brain Neoplasms/pathology , Brain Neoplasms/therapy , Cancer Vaccines/administration & dosage , Cytokines/therapeutic use , Dendritic Cells/immunology , Dendritic Cells/transplantation , Glioblastoma/immunology , Glioblastoma/pathology , Glioblastoma/therapy , Humans , Immunotherapy/methods , Neoplastic Cells, Circulating/immunology , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/transplantation
8.
Cytotherapy ; 16(8): 1121-31, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24841514

ABSTRACT

BACKGROUND AIMS: Outcomes for patients with glioblastoma remain poor despite aggressive multimodal therapy. Immunotherapy with genetically modified T cells expressing chimeric antigen receptors (CARs) targeting interleukin (IL) 13Rα2, human epidermal growth factor receptor 2, epidermal growth factor variant III or erythropoietin-producing hepatocellular carcinoma A2 has shown promise for the treatment of glioma in preclinical models. On the basis of IL13Rα2 immunotoxins that contain IL13 molecules with one or two amino acid substitutions (IL13 muteins) to confer specificity to IL13Rα2, investigators have constructed CARS with IL13 muteins as antigen-binding domains. Whereas the specificity of IL13 muteins in the context of immunotoxins is well characterized, limited information is available for CAR T cells. METHODS: We constructed four second-generation CARs with IL13 muteins with one or two amino acid substitutions, and evaluated the effector function of IL13-mutein CAR T cells in vitro and in vivo. RESULTS: T cells expressing all four CARs recognized IL13Rα1 or IL13Rα2 recombinant protein in contrast to control protein (IL4R) as judged by interferon-γ production. IL13 protein produced significantly more IL2, indicating that IL13 mutein-CAR T cells have a higher affinity to IL13Rα2 than to IL13Rα1. In cytotoxicity assays, CAR T cells killed IL13Rα1- and/or IL13Rα2-positive cells in contrast to IL13Rα1- and IL13Rα2-negative controls. Although we observed no significant differences between IL13 mutein-CAR T cells in vitro, only T cells expressing IL13 mutein-CARs with an E13K amino acid substitution had anti-tumor activity in vivo that resulted in a survival advantage of treated animals. CONCLUSIONS: Our study highlights that the specificity/avidity of ligands is context-dependent and that evaluating CAR T cells in preclinical animal model is critical to assess their potential benefit.


Subject(s)
Glioblastoma/therapy , Immunotherapy , Interleukin-13 Receptor alpha2 Subunit/genetics , T-Lymphocytes/immunology , Amino Acid Substitution , Animals , Gene Expression Regulation, Neoplastic/immunology , Glioblastoma/immunology , Glioblastoma/pathology , Humans , Immunotoxins/genetics , Interleukin-13 Receptor alpha1 Subunit/genetics , Interleukin-13 Receptor alpha1 Subunit/immunology , Interleukin-13 Receptor alpha2 Subunit/immunology , Mice , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Xenograft Model Antitumor Assays
9.
Mol Ther ; 21(11): 2087-101, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23939024

ABSTRACT

Preclinical and early clinical studies have demonstrated that chimeric antigen receptor (CAR)-redirected T cells are highly promising in cancer therapy. We observed that targeting HER2 in a glioblastoma (GBM) cell line results in the emergence of HER2-null tumor cells that maintain the expression of nontargeted tumor-associated antigens. Combinational targeting of these tumor-associated antigens could therefore offset this escape mechanism. We studied the single-cell coexpression patterns of HER2, IL-13Rα2, and EphA2 in primary GBM samples using multicolor flow cytometry and immunofluorescence, and applied a binomial routine to the permutations of antigen expression and the related odds of complete tumor elimination. This mathematical model demonstrated that cotargeting HER2 and IL-13Rα2 could maximally expand the therapeutic reach of the T cell product in all primary tumors studied. Targeting a third antigen did not predict an added advantage in the tumor cohort studied. We therefore generated bispecific T cell products from healthy donors and from GBM patients by pooling T cells individually expressing HER2 and IL-13Rα2-specific CARs and by making individual T cells to coexpress both molecules. Both HER2/IL-13Rα2-bispecific T cell products offset antigen escape, producing enhanced effector activity in vitro immunoassays (against autologous glioma cells in the case of GBM patient products) and in an orthotopic xenogeneic murine model. Further, T cells coexpressing HER2 and IL-13Rα2-CARs exhibited accentuated yet antigen-dependent downstream signaling and a particularly enhanced antitumor activity.


Subject(s)
Adoptive Transfer , Antigens, Neoplasm/metabolism , Glioblastoma/therapy , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/immunology , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Cell Line, Tumor , Combined Modality Therapy , Glioblastoma/immunology , Glioblastoma/pathology , HEK293 Cells , Humans , Interleukin-13 Receptor alpha2 Subunit/genetics , Interleukin-13 Receptor alpha2 Subunit/immunology , Interleukin-13 Receptor alpha2 Subunit/metabolism , Mice , Mice, SCID , Models, Biological , Receptor, ErbB-2/genetics , Receptor, ErbB-2/immunology , Receptor, ErbB-2/metabolism , Receptors, Antigen, T-Cell/immunology , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , Tumor Cells, Cultured , Tumor Escape , Xenograft Model Antitumor Assays
10.
Mol Ther ; 21(8): 1611-20, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23732988

ABSTRACT

Cancer-associated fibroblasts (CAFs), the principle component of the tumor-associated stroma, form a highly protumorigenic and immunosuppressive microenvironment that mediates therapeutic resistance. Co-targeting CAFs in addition to cancer cells may therefore augment the antitumor response. Fibroblast activation protein-α (FAP), a type 2 dipeptidyl peptidase, is expressed on CAFs in a majority of solid tumors making it an attractive immunotherapeutic target. To target FAP-positive CAFs in the tumor-associated stroma, we genetically modified T cells to express a FAP-specific chimeric antigen receptor (CAR). The resulting FAP-specific T cells recognized and killed FAP-positive target cells as determined by proinflammatory cytokine release and target cell lysis. In an established A549 lung cancer model, adoptive transfer of FAP-specific T cells significantly reduced FAP-positive stromal cells, with a concomitant decrease in tumor growth. Combining these FAP-specific T cells with T cells that targeted the EphA2 antigen on the A549 cancer cells themselves significantly enhanced overall antitumor activity and conferred a survival advantage compared to either alone. Our study underscores the value of co-targeting both CAFs and cancer cells to increase the benefits of T-cell immunotherapy for solid tumors.


Subject(s)
Fibroblasts/immunology , Gelatinases/immunology , Membrane Proteins/immunology , Neoplasms/immunology , Receptors, Antigen/immunology , Serine Endopeptidases/immunology , T-Lymphocytes/immunology , Animals , Cell Line, Tumor , Cytokines/biosynthesis , Cytotoxicity, Immunologic , Disease Models, Animal , Endopeptidases , Fibroblasts/metabolism , Gelatinases/genetics , Gelatinases/metabolism , Gene Expression , Gene Order , Genetic Vectors , Humans , Immunotherapy , Inflammation Mediators/metabolism , Lung/immunology , Lung/metabolism , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Neoplasms/metabolism , Neoplasms/mortality , Neoplasms/pathology , Receptors, Antigen/genetics , Receptors, Antigen/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , T-Lymphocytes/metabolism
11.
Mol Ther ; 21(3): 629-37, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23070117

ABSTRACT

Outcomes for patients with glioblastoma (GBM) remain poor despite aggressive multimodal therapy. Immunotherapy with genetically modified T cells expressing chimeric antigen receptors (CARs) targeting interleukin (IL)-13Rα2, epidermal growth factor receptor variant III (EGFRvIII), or human epidermal growth factor receptor 2 (HER2) has shown promise for the treatment of gliomas in preclinical models and in a clinical study (IL-13Rα2). However, targeting IL-13Rα2 and EGFRvIII is associated with the development of antigen loss variants, and there are safety concerns with targeting HER2. Erythropoietin-producing hepatocellular carcinoma A2 (EphA2) has emerged as an attractive target for the immunotherapy of GBM as it is overexpressed in glioma and promotes its malignant phenotype. To generate EphA2-specific T cells, we constructed an EphA2-specific CAR with a CD28-ζ endodomain. EphA2-specific T cells recognized EphA2-positive glioma cells as judged by interferon-γ (IFN-γ) and IL-2 production and tumor cell killing. In addition, EphA2-specific T cells had potent activity against human glioma-initiating cells preventing neurosphere formation and destroying intact neurospheres in coculture assays. Adoptive transfer of EphA2-specific T cells resulted in the regression of glioma xenografts in severe combined immunodeficiency (SCID) mice and a significant survival advantage in comparison to untreated mice and mice treated with nontransduced T cells. Thus, EphA2-specific T-cell immunotherapy may be a promising approach for the treatment of EphA2-positive GBM.


Subject(s)
Genetic Therapy/methods , Glioblastoma/therapy , Immunotherapy/methods , Receptor, EphA2/genetics , T-Lymphocytes/immunology , Adoptive Transfer , Animals , Cell Line, Tumor , Genetic Vectors , Humans , Interferon-gamma/metabolism , Interleukin-2/metabolism , K562 Cells , Male , Mice , Mice, SCID , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Retroviridae/genetics , Transduction, Genetic
12.
PLoS One ; 6(5): e20267, 2011.
Article in English | MEDLINE | ID: mdl-21647415

ABSTRACT

Cancer cells can live and grow if they succeed in creating a favorable niche that often includes elements from the immune system. While T lymphocytes play an important role in the host response to tumor growth, the mechanism of their trafficking to the tumor remains poorly understood. We show here that T lymphocytes consistently infiltrate the primary brain cancer, medulloblastoma. We demonstrate, both in vitro and in vivo, that these T lymphocytes are attracted to tumor deposits only after the tumor cells have interacted with tumor vascular endothelium. Macrophage Migration Inhibitory Factor (MIF)" is the key chemokine molecule secreted by tumor cells which induces the tumor vascular endothelial cells to secrete the potent T lymphocyte attractant "Regulated upon Activation, Normal T-cell Expressed, and Secreted (RANTES)." This in turn creates a chemotactic gradient for RANTES-receptor bearing T lymphocytes. Manipulation of this pathway could have important therapeutic implications.


Subject(s)
Cerebellar Neoplasms/pathology , Chemotaxis , Endothelial Cells/pathology , Medulloblastoma/pathology , Signal Transduction , T-Lymphocytes/immunology , Tumor Microenvironment , Animals , Brain/immunology , Brain/metabolism , Brain/pathology , Cerebellar Neoplasms/immunology , Cerebellar Neoplasms/metabolism , Chemokine CCL5/metabolism , Endothelial Cells/immunology , Endothelial Cells/metabolism , Humans , Lymphocytes, Tumor-Infiltrating/cytology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Macrophage Migration-Inhibitory Factors/metabolism , Male , Medulloblastoma/immunology , Medulloblastoma/metabolism , Mice , T-Lymphocytes/cytology , T-Lymphocytes/metabolism
13.
Blood ; 117(16): 4304-14, 2011 Apr 21.
Article in English | MEDLINE | ID: mdl-21304103

ABSTRACT

T-cell therapy with genetically modified T cells targeting CD19 or CD20 holds promise for the immunotherapy of hematologic malignancies. These targets, however, are only present on B cell-derived malignancies, and because they are broadly expressed in the hematopoietic system, their targeting may have unwanted consequences. To expand T-cell therapies to hematologic malignancies that are not B cell-derived, we determined whether T cells can be redirected to CD70, an antigen expressed by limited subsets of normal lymphocytes and dendritic cells, but aberrantly expressed by a broad range of hematologic malignancies and some solid tumors. To generate CD70-specific T cells, we constructed a chimeric antigen receptor (CAR) consisting of the CD70 receptor (CD27) fused to the CD3-ζ chain. Stimulation of T cells expressing CD70-specific CARs resulted in CD27 costimulation and recognition of CD70-positive tumor cell lines and primary tumor cells, as shown by IFN-γ and IL-2 secretion and by tumor cell killing. Adoptively transferred CD70-specific T cells induced sustained regression of established murine xenografts. Therefore, CD70-specific T cells may be a promising immunotherapeutic approach for CD70-positive malignancies.


Subject(s)
CD27 Ligand/immunology , Immunotherapy , Lymphoma, Non-Hodgkin/therapy , T-Lymphocytes/immunology , Animals , Cell Line, Tumor , Cell Proliferation , Cell Survival , Cells, Cultured , Cytokines/immunology , Humans , Immunotherapy/methods , Interleukin-2/immunology , Lymphocyte Activation , Lymphoma, Non-Hodgkin/immunology , Mice , Mice, SCID , T-Lymphocytes/transplantation , Tumor Necrosis Factor Receptor Superfamily, Member 7/immunology
SELECTION OF CITATIONS
SEARCH DETAIL