Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
3.
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
4.
J Clin Oncol ; 33(15): 1688-96, 2015 May 20.
Article in English | MEDLINE | ID: mdl-25800760

ABSTRACT

PURPOSE: The outcome for patients with metastatic or recurrent sarcoma remains poor. Adoptive therapy with tumor-directed T cells is an attractive therapeutic option but has never been evaluated in sarcoma. PATIENTS AND METHODS: We conducted a phase I/II clinical study in which patients with recurrent/refractory human epidermal growth factor receptor 2 (HER2) -positive sarcoma received escalating doses (1 × 10(4)/m(2) to 1 × 10(8)/m(2)) of T cells expressing an HER2-specific chimeric antigen receptor with a CD28.ζ signaling domain (HER2-CAR T cells). RESULTS: We enrolled 19 patients with HER2-positive tumors (16 osteosarcomas, one Ewing sarcoma, one primitive neuroectodermal tumor, and one desmoplastic small round cell tumor). HER2-CAR T-cell infusions were well tolerated with no dose-limiting toxicity. At dose level 3 (1 × 10(5)/m(2)) and above, we detected HER2-CAR T cells 3 hours after infusion by quantitative polymerase chain reaction in 14 of 16 patients. HER2-CAR T cells persisted for at least 6 weeks in seven of the nine evaluable patients who received greater than 1 × 10(6)/m(2) HER2-CAR T cells (P = .005). HER2-CAR T cells were detected at tumor sites of two of two patients examined. Of 17 evaluable patients, four had stable disease for 12 weeks to 14 months. Three of these patients had their tumor removed, with one showing ≥ 90% necrosis. The median overall survival of all 19 infused patients was 10.3 months (range, 5.1 to 29.1 months). CONCLUSION: This first evaluation of the safety and efficacy of HER2-CAR T cells in patients with cancer shows the cells can persist for 6 weeks without evident toxicities, setting the stage for studies that combine HER2-CAR T cells with other immunomodulatory approaches to enhance their expansion and persistence.


Subject(s)
Bone Neoplasms/therapy , Immunotherapy/methods , Receptor, ErbB-2/metabolism , Sarcoma/therapy , T-Lymphocytes/immunology , Adolescent , Adult , Bone Neoplasms/metabolism , Child , Female , Humans , Kaplan-Meier Estimate , Magnetic Resonance Imaging , Male , Maximum Tolerated Dose , Neoplasm Metastasis , Neuroectodermal Tumors/metabolism , Neuroectodermal Tumors/therapy , Osteosarcoma/metabolism , Osteosarcoma/therapy , Positron-Emission Tomography , Receptor, ErbB-2/genetics , Receptors, Antigen, T-Cell/chemistry , Recurrence , Sarcoma/metabolism , Sarcoma, Ewing/metabolism , Sarcoma, Ewing/therapy , Tomography, X-Ray Computed , Treatment Outcome , Young Adult
5.
J Neurooncol ; 119(1): 27-35, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24792645

ABSTRACT

Separase, an enzyme that cleaves the chromosomal cohesin during mitosis, is overexpressed in a wide range of human epithelial cancers of breast, bone and prostate (Meyer et al., Clin Cancer Res 15(8):2703-2710, 2009). Overexpression of Separase in animal models results in aneuploidy and tumorigenesis. We have examined the expression and localization of Separase protein in adult and pediatric glioblastoma and normal brain specimens. Immunofluorescence microscopy and Western blot analysis showed significant overexpression of Separase in all adult and a subset of pediatric glioblastoma cells. Tumor status and patient survival strongly correlate with the mislocalization of Separase into the nucleus throughout all stages of the cell cycle. Unlike exclusively nuclear localization in mitotic control cells, glioblastoma samples have a significantly higher number of resting (interphase) cells with strong nuclear Separase staining. Additionally, patient survival analysis demonstrated a strong correlation between overexpression of Separase protein in adult glioblastoma and a high incidence of relapse and reduced overall survival. These results further strengthen our hypothesis that Separase is an oncogene whose overexpression induces tumorigenesis, and indicate that Separase overexpression and aberrant nuclear localization are common in many tumor types and may predict outcome in some human malignancies.


Subject(s)
Brain Neoplasms/metabolism , Cell Nucleus/metabolism , Glioblastoma/metabolism , Separase/metabolism , Up-Regulation , Brain Neoplasms/mortality , Cell Cycle , Glioblastoma/mortality , Humans , Prognosis , Recurrence , Survival Rate
6.
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
7.
J Neurosci ; 33(33): 13560-8, 2013 Aug 14.
Article in English | MEDLINE | ID: mdl-23946414

ABSTRACT

Contemporary views of tumorigenesis regard its inception as a convergence of genetic mutation and developmental context. Glioma is the most common and deadly malignancy in the CNS; therefore, understanding how regulators of glial development contribute to its formation remains a key question. Previously we identified nuclear factor I-A (NFIA) as a key regulator of developmental gliogenesis, while miR-223 has been shown to repress NFIA expression in other systems. Using this relationship as a starting point, we found that miR-223 can suppress glial precursor proliferation via repression of NFIA during chick spinal cord development. This relationship is conserved in glioma, as miR-223 and NFIA expression is negatively correlated in human glioma tumors, and the miR-223/NFIA axis suppresses tumorigenesis in a human glioma cell line. Subsequent analysis of NFIA function revealed that it directly represses p21 and is required for tumorigenesis in a mouse neural stem cell model of glioma. These studies represent the first characterization of miR-223/NFIA axis function in glioma and demonstrate that it is a conserved proliferative mechanism across CNS development and tumorigenesis.


Subject(s)
Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Glioma/metabolism , MicroRNAs/metabolism , NFI Transcription Factors/metabolism , Neoplastic Stem Cells/metabolism , Animals , Cell Transformation, Neoplastic/genetics , Chick Embryo , Chromatin Immunoprecipitation , Gene Expression Regulation, Neoplastic/physiology , Glioma/genetics , Glioma/pathology , Humans , Immunohistochemistry , In Situ Hybridization , Mice , MicroRNAs/genetics , NFI Transcription Factors/genetics , Neoplastic Stem Cells/pathology , Neuroglia/metabolism , Neuroglia/pathology , Rats , Reverse Transcriptase Polymerase Chain Reaction , Tissue Array Analysis , Xenograft Model Antitumor Assays
8.
Clin Immunol ; 149(1): 55-64, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23891738

ABSTRACT

Medulloblastoma (MB) is the most common malignant brain tumor of childhood. Current therapies are toxic and not always curative that necessitates development of targeted immunotherapy. However, little is known about immunobiology of this tumor. In this study, we show that MB cells in 9 of 20 primary tumors express CD1d, an antigen-presenting molecule for Natural Killer T cells (NKTs). Quantitative RT-PCR analysis of 61 primary tumors revealed an elevated level of CD1d mRNA expression in a molecular subgroup characterized by an overactivation of Sonic Hedgehog (SHH) oncogene compared with Group 4. CD1d-positive MB cells cross-presented glycolipid antigens to activate NKT-cell cytotoxicity. Intracranial injection of NKTs resulted in regression of orthotopic MB xenografts in NOD/SCID mice. Importantly, the numbers and function of peripheral blood type-I NKTs were preserved in MB patients. Therefore, CD1d is expressed on tumor cells in a subset of MB patients and represents a novel target for immunotherapy.


Subject(s)
Antigens, CD1d/immunology , Immunotherapy , Medulloblastoma/therapy , Natural Killer T-Cells/immunology , Adolescent , Animals , Antigens, CD1d/genetics , Cell Line, Tumor , Child , Child, Preschool , Humans , Male , Medulloblastoma/immunology , Mice , Mice, SCID , RNA, Messenger/metabolism
9.
Mol Ther Nucleic Acids ; 2: e105, 2013 Jul 09.
Article in English | MEDLINE | ID: mdl-23839099

ABSTRACT

Targeted T cells are emerging as effective non-toxic therapies for cancer. Multiple elements, however, contribute to the overall pathogenesis of cancer through both distinct and redundant mechanisms. Hence, targeting multiple cancer-specific markers simultaneously could result in better therapeutic efficacy. We created a functional chimeric antigen receptor-the TanCAR, a novel artificial molecule that mediates bispecific activation and targeting of T cells. We demonstrate the feasibility of cumulative integration of structure and docking simulation data using computational tools to interrogate the design and predict the functionality of such a complex bispecific molecule. Our prototype TanCAR induced distinct T cell reactivity against each of two tumor restricted antigens, and produced synergistic enhancement of effector functions when both antigens were simultaneously encountered. Furthermore, the TanCAR preserved the cytolytic ability of T cells upon loss of one of the target molecules and better controlled established experimental tumors by recognition of both targets in an animal disease model. This proof-of-concept approach can be used to increase the specificity of effector cells for malignant versus normal target cells, to offset antigen escape or to allow for targeting the tumor and its microenvironment.Molecular Therapy-Nucleic Acids (2013) 2, e105; doi:10.1038/mtna.2013.32; published online 9 July 2013.

10.
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
11.
J Immunother ; 35(2): 159-68, 2012.
Article in English | MEDLINE | ID: mdl-22306904

ABSTRACT

Glioblastoma (GBM) is the most common primary brain cancer in adults and is virtually incurable. Recent studies have shown that cytomegalovirus (CMV) is present in majority of GBMs. To evaluate whether the CMV antigens pp65 and IE1, which are expressed in GBMs, could be targeted by CMV-specific T cells, we measured the frequency of T cells targeting pp65 and IE1 in the peripheral blood of a cohort of 11 sequentially diagnosed CMV-seropositive GBM patients, and evaluated whether it was feasible to expand autologous CMV-specific T cells for future clinical studies. All 11 CMV-seropositive GBM patients had T cells specific for pp65 and IE1 in their peripheral blood assessed by IFNγ enzyme-linked immunospot assay. However, the precursor frequency of pp65-specific T cells was decreased in comparison with healthy donors (P=0.001). We successfully reactivated and expanded CMV-specific T cells from 6 out of 6 GBM patients using antigen-presenting cells transduced with an adenoviral vector encoding pp65 and IE1. CMV-specific T-cell lines contained CD4 as well as CD8 T cells, recognized pp65 and IE1 targets and killed CMV-infected autologous GBM cells. Infusion of such CMV-specific T-cell lines may extend the benefits of T-cell therapy to patients with CMV GBMs.


Subject(s)
Antigens, Viral/immunology , Brain Neoplasms/therapy , Glioblastoma/therapy , Immunotherapy, Adoptive/methods , T-Lymphocytes/immunology , Adult , Aged , Brain Neoplasms/immunology , Brain Neoplasms/virology , Cell Separation , Cytomegalovirus/immunology , Cytomegalovirus Infections/immunology , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Glioblastoma/immunology , Glioblastoma/virology , Humans , In Situ Hybridization , Male , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL
...