Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
2.
J Hematol Oncol ; 11(1): 92, 2018 07 06.
Article in English | MEDLINE | ID: mdl-29980239

ABSTRACT

BACKGROUND: Triple-negative breast cancer (TNBC) is an aggressive disease that currently lacks effective targeted therapy. NKG2D ligands (NKG2DLs) are expressed on various tumor types and immunosuppressive cells within tumor microenvironments, providing suitable targets for cancer therapy. METHODS: We applied a chimeric antigen receptor (CAR) approach for the targeting of NKG2DLs expressed on human TNBCs. Lentiviral vectors were used to express the extracellular domain of human NKG2D that binds various NKG2DLs, fused to signaling domains derived from T cell receptor CD3 zeta alone or with CD27 or 4-1BB (CD137) costimulatory domain. RESULTS: Interleukin-2 (IL-2) promoted the expansion and self-enrichment of NKG2D-redirected CAR T cells in vitro. High CD25 expression on first-generation NKG2D CAR T cells was essential for the self-enrichment effect in the presence of IL-2, but not for CARs containing CD27 or 4-1BB domains. Importantly, self-enriched NKG2D CAR T cells effectively recognized and eliminated TNBC cell lines in vitro, and adoptive transfer of T cells expressing NKG2D CARs with CD27 or 4-1BB specifically enhanced NKG2D CAR surface expression, T cell persistence, and the regression of established MDA-MB-231 TNBC in vivo. NKG2D-z CAR T cells lacking costimulatory domains were less effective, highlighting the need for costimulatory signals. CONCLUSIONS: These results demonstrate that CD27 or 4-1BB costimulated, self-enriched NKG2D CAR-redirected T cells mediate anti-tumor activity against TNBC tumor, which represent a promising immunotherapeutic approach to TNBC treatment.


Subject(s)
Immunotherapy/methods , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Receptors, Chimeric Antigen/therapeutic use , Triple Negative Breast Neoplasms/therapy , Female , Humans , Transfection , Triple Negative Breast Neoplasms/pathology
3.
Oncotarget ; 7(50): 82354-82368, 2016 Dec 13.
Article in English | MEDLINE | ID: mdl-27409425

ABSTRACT

Exogenous cytokines are widely applied to enhance the anti-tumor ability of immune cells. However, systematic comparative studies of their effects on chimeric antigen receptor (CAR)-engineered T (CART) cells are lacking. In this study, CART cells targeting folate receptor-alpha were generated and expanded ex vivo in the presence of different cytokines (IL-2, IL-7, IL-15, IL-18, and IL-21), and their expansion, phenotype and cytotoxic capacity were evaluated, in vitro and in vivo. Moreover, the effect of the administration of these cytokines along with CART cells in vivo was also studied. IL-2, IL-7, and IL-15 favored the ex vivo expansion of CART cells compared to other cytokines or no cytokine treatment. IL-7 induced the highest proportion of memory stem cell-like CART cells in the final product, and IL-21 supported the expansion of CART cells with a younger phenotype, while IL-2 induced more differentiated CART cells. IL-2 and IL-15-exposed CART cells secreted more proinflammatory cytokines and presented stronger tumor-lysis ability in vitro. However, when tested in vivo, CART cells exposed to IL-2 ex vivo showed the least anti-tumor effect. In contrast, the administration of IL-15 and IL-21 in combination with CART cells in vivo increased their tumor killing capacity. According to our results, IL-7 and IL-15 show promise to promote ex vivo expansion of CART cells, while IL-15 and IL-21 seem better suited for in vivo administration after CART cell infusion. Collectively, these results may have a profound impact on the efficacy of CART cells in both hematologic and solid cancers.


Subject(s)
Cytokines/pharmacology , Cytotoxicity, Immunologic/drug effects , Immunotherapy, Adoptive , Lymphocyte Activation/drug effects , Ovarian Neoplasms/therapy , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/transplantation , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Coculture Techniques , Female , Humans , Mice, Inbred NOD , Mice, SCID , Ovarian Neoplasms/immunology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Phenotype , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Recombinant Fusion Proteins/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Time Factors , Transfection , Xenograft Model Antitumor Assays
4.
J Hematol Oncol ; 9(1): 56, 2016 07 20.
Article in English | MEDLINE | ID: mdl-27439908

ABSTRACT

BACKGROUND: The poor prognosis and the limited efficacy of targeted therapy in patients with triple-negative breast cancer (TNBC) have raised the need for alternative therapies. Recent studies have demonstrated that folate receptor-alpha (FRα) may represent an ideal tumor-associated marker for immunotherapy for TNBC. METHODS: The aim of the present study was to apply a chimeric antigen receptor (CAR) approach for the targeting of FRα expressed on TNBC cells and evaluate the antitumor activity of CAR-engineered T cells in vitro and in vivo. RESULTS: We found that human T cells expressing a FRα-specific CAR were potent and specific killers of TNBC cells that express moderate levels of FRα in vitro and significantly inhibited tumor outgrowth following infusion into immunodeficient mice bearing an MDA-MB-231 tumor xenograft. However, the antitumor activity of the FRα CAR T cells was modest when compared to the same CAR T cells applied in an ovarian tumor xenograft model where FRα expression is more abundant. Notably, FRα CAR T cells induced superior tumor regression in vivo against MDA-MB-231 that was engineered for overexpression of FRα. CONCLUSIONS: Taken together, our results show that FRα CAR T cells can mediate antitumor activity against established TNBC tumor, particularly when FRα is expressed at higher levels. These results have significant implications for the pre-selection of patients with high antigen expression levels when utilizing CAR-based adoptive T cell therapies of cancer in future clinical trials.


Subject(s)
Folate Receptor 1/analysis , Immunotherapy, Adoptive/methods , Ovarian Neoplasms/therapy , Triple Negative Breast Neoplasms/therapy , Animals , Cells, Cultured , Female , Folate Receptor 1/immunology , Heterografts , Humans , Immunotherapy, Adoptive/standards , Mice , Ovarian Neoplasms/chemistry , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes/immunology , T-Lymphocytes/transplantation , Triple Negative Breast Neoplasms/chemistry
5.
Oncotarget ; 6(25): 21533-46, 2015 Aug 28.
Article in English | MEDLINE | ID: mdl-26101914

ABSTRACT

Chimeric antigen receptors (CARs) can redirect T cells against antigen-expressing tumors in an HLA-independent manner. To date, various CARs have been constructed using mouse single chain antibody variable fragments (scFvs) of high affinity that are immunogenic in humans and have the potential to mediate "on-target" toxicity. Here, we developed and evaluated a fully human CAR comprised of the human C4 folate receptor-alpha (αFR)-specific scFv coupled to intracellular T cell signaling domains. Human T cells transduced to express the C4 CAR specifically secreted proinflammatory cytokine and exerted cytolytic functions when cultured with αFR-expressing tumors in vitro. Adoptive transfer of C4 CAR T cells mediated the regression of large, established human ovarian cancer in a xenogeneic mouse model. Relative to a murine MOv19 scFv-based αFR CAR, C4 CAR T cells mediated comparable cytotoxic tumor activity in vitro and in vivo but had lower affinity for αFR protein and exhibited reduced recognition of normal cells expressing low levels of αFR. Thus, T cells expressing a fully human CAR of intermediate affinity can efficiently kill antigen-expressing tumors in vitro and in vivo and may overcome issues of transgene immunogenicity and "on-target off-tumor" toxicity that plague trials utilizing CARs containing mouse-derived, high affinity scFvs.


Subject(s)
Folate Receptor 1/chemistry , HLA Antigens/chemistry , Neoplasm Transplantation , Ovarian Neoplasms/therapy , Receptors, Antigen/chemistry , Amino Acid Motifs , Animals , CD4-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/cytology , Cell Separation , Cytokines/metabolism , Female , Flow Cytometry , HEK293 Cells , Humans , Immunotherapy, Adoptive , Inflammation , Lentivirus/metabolism , Mice , Mice, Nude , Mice, SCID , Ovarian Neoplasms/metabolism , Protein Structure, Tertiary , Signal Transduction , Tumor Necrosis Factor Receptor Superfamily, Member 7/chemistry , Xenograft Model Antitumor Assays
6.
Clin Cancer Res ; 20(1): 44-55, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24045181

ABSTRACT

PURPOSE: Upregulation of CD137 (4-1BB) on recently activated CD8(+) T cells has been used to identify rare viral or tumor antigen-specific T cells from peripheral blood. Here, we evaluated the immunobiology of CD137 in human cancer and the utility of a CD137-positive separation methodology for the identification and enrichment of fresh tumor-reactive tumor-infiltrating lymphocytes (TIL) or tumor-associated lymphocytes (TAL) from ascites for use in adoptive immunotherapy. EXPERIMENTAL DESIGN: TILs from resected ovarian cancer or melanoma were measured for surface CD137 expression directly or after overnight incubation in the presence of tumor cells and homeostatic cytokines. CD137(pos) TILs were sorted and evaluated for antitumor activity in vitro and in vivo. RESULTS: Fresh ovarian TILs and TALs naturally expressed higher levels of CD137 than circulating T cells. An HLA-dependent increase in CD137 expression was observed following incubation of fresh enzyme-digested tumor or ascites in IL-7 and IL-15 cytokines, but not IL-2. Enriched CD137(pos) TILs, but not PD-1(pos) or PD-1(neg) CD137(neg) cells, possessed autologous tumor reactivity in vitro and in vivo. In melanoma studies, all MART-1-specific CD8(+) TILs upregulated CD137 expression after incubation with HLA-matched, MART-expressing cancer cells and antigen-specific effector function was restricted to the CD137(pos) subset in vitro. CD137(pos) TILs also mediated superior antitumor effects in vivo, compared with CD137(neg) TILs. CONCLUSIONS: Our findings reveal a role for the TNFR-family member CD137 in the immunobiology of human cancer where it is preferentially expressed on tumor-reactive subset of TILs, thus rationalizing its agonistic engagement in vivo and its use in TIL selection for adoptive immunotherapy trials.


Subject(s)
Melanoma/immunology , Ovarian Neoplasms/immunology , T-Lymphocytes/metabolism , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism , Animals , Cell Line, Tumor , Cell Separation , Coculture Techniques , Female , Gene Expression , Gene Expression Regulation, Neoplastic , Humans , Leukocytes, Mononuclear/metabolism , Melanoma/metabolism , Melanoma/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Tumor Burden , Tumor Necrosis Factor Receptor Superfamily, Member 9/genetics , Up-Regulation
7.
Hum Gene Ther ; 24(3): 295-305, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23297870

ABSTRACT

NKG2D ligands (NKG2DLs) are widely expressed on ovarian cancers to various degrees, making them attractive targets for immunotherapy. Here, we applied a chimeric antigen receptor (CAR) approach for the targeting of NKG2DLs expressed on human ovarian cancer cells and evaluated the impact of pharmacological upregulation of NKG2DLs on immune recognition. Various NKG2DLs, including MICA/B and ULBP-1, -2, -3, and -4, were expressed at various levels on the surface of all established ovarian cancer cell lines and primary ovarian cancer samples tested. To redirect human T cells against NKG2DLs, an NKG2DL-specific CAR was generated by fusing the extracellular domain of the NKG2D receptor to the 4-1BB costimulatory and CD3-ζ chain signaling domains. In vitro expansion of chimeric NKG2D CAR T cells was delayed compared with untransduced T cells and control CAR T cells; the likely result of fratricide among activated T cells expressing NKG2DLs. However, NKG2D CAR T cells did expand and were selectively enriched during prolonged culture. In coculture, CD4(+) and CD8(+) NKG2D CAR T cells specifically recognized and killed NKG2DL-expressing ovarian cancer cell lines but not NKG2DL-negative cells. Notably, pretreatment of ovarian cancer cells expressing moderate to low levels of NKG2DLs with the histone deacetylase inhibitor sodium valproate (VPA) upregulated NKG2DL cell surface expression and consequently enhanced their immune recognition by chimeric NKG2D CAR T cells. Our results demonstrate that VPA-induced upregulation of NKG2DL expression enhances the immune recognition of ovarian cancer cells by engineered NKG2D CAR T cells, and rationalizes the use of VPA in combination with NKG2DL-targeted immunotherapy in ovarian cancer.


Subject(s)
Histone Deacetylases/metabolism , NK Cell Lectin-Like Receptor Subfamily K/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/immunology , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Cell Line , Female , Gene Expression , Gene Expression Regulation, Neoplastic/drug effects , Gene Order , Genetic Vectors/genetics , Histone Deacetylase Inhibitors/pharmacology , Humans , Immunophenotyping , Lentivirus/genetics , Ligands , Lymphocyte Activation , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Ovarian Neoplasms/metabolism , Phenotype , Receptors, Antigen, T-Cell/metabolism , Valproic Acid/pharmacology
8.
Oncoimmunology ; 2(12): e27184, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24501691

ABSTRACT

We have recently identified tumor necrosis factor receptor superfamily, member 9 (TNFRSF9, best known as CD137 or 4-1BB) as a biomarker of tumor-reactive T cells naturally occurring in cancer patients, and developed a rapid, accurate system to comprehensively isolate lymphocytes with tumor-rejecting properties from human biopsies. Our findings reveal a previously unappreciated role for CD137, a co-stimulatory TNFR family member, in the immunobiology of human cancer.

9.
PLoS One ; 7(11): e49829, 2012.
Article in English | MEDLINE | ID: mdl-23189165

ABSTRACT

The breadth of HER2 expression by primary human ovarian cancers remains controversial, which questions its suitability as a universal antigen in this malignancy. To address these issues, we performed extensive HER2 expression analysis on a wide panel of primary tumors as well as established and short-term human ovarian cancer cell lines. Conventional immunohistochemical (IHC) analysis of multiple tumor sites in 50 cases of high-grade ovarian serous carcinomas revealed HER2 overexpression in 29% of evaluated sites. However, more sensitive detection methods including flow cytometry, western blot analysis and q-PCR revealed HER2 expression in all fresh tumor cells derived from primary ascites or solid tumors as well as all established and short-term cultured cancer cell lines. Cancer cells generally expressed HER2 at higher levels than that found in normal ovarian surface epithelial (OSE) cells. Accordingly, genetically-engineered human T cells expressing an HER2-specific chimeric antigen receptor (CAR) recognized and reacted against all established or primary ovarian cancer cells tested with minimal or no reactivity against normal OSE cells. In conclusion, all human ovarian cancers express immunologically-detectable levels of HER2, indicating that IHC measurement underestimates the true frequency of HER2-expressing ovarian cancers and may limit patient access to otherwise clinically meaningful HER2-targeted therapies.


Subject(s)
Neoplasms, Glandular and Epithelial/metabolism , Ovarian Neoplasms/metabolism , Receptor, ErbB-2/metabolism , Carcinoma, Ovarian Epithelial , Cell Line, Tumor , Epithelial Cells/metabolism , Female , Gene Expression , Humans , Immunohistochemistry , Neoplasms, Glandular and Epithelial/genetics , Ovarian Neoplasms/genetics , Ovary/cytology , Ovary/metabolism , Receptor, ErbB-2/genetics , Receptor, ErbB-2/immunology , T-Lymphocytes/immunology
10.
Oncoimmunology ; 1(4): 547-549, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22754782

ABSTRACT

The impact of CD27 costimulation on human T lymphocyte effector function and memory formation has been limited to evaluations using in vitro cell culture systems and observations from the clinic. We therefore investigated whether CD27 signaling augments antigen redirected human T-cell survival, expansion and function in vitro and in vivo.

11.
Blood ; 119(3): 696-706, 2012 Jan 19.
Article in English | MEDLINE | ID: mdl-22117050

ABSTRACT

The costimulatory effects of CD27 on T lymphocyte effector function and memory formation has been confined to evaluations in mouse models, in vitro human cell culture systems, and clinical observations. Here, we tested whether CD27 costimulation actively enhances human T-cell function, expansion, and survival in vitro and in vivo. Human T cells transduced to express an antigen-specific chimeric antigen receptor (CAR-T) containing an intracellular CD3 zeta (CD3ζ) chain signaling module with the CD27 costimulatory motif in tandem exerted increased antigen-stimulated effector functions in vitro, including cytokine secretion and cytotoxicity, compared with CAR-T with CD3ζ alone. After antigen stimulation in vitro, CD27-bearing CAR-T cells also proliferated, up-regulated Bcl-X(L) protein expression, resisted apoptosis, and underwent increased numerical expansion. The greatest impact of CD27 was noted in vivo, where transferred CAR-T cells with CD27 demonstrated heightened persistence after infusion, facilitating improved regression of human cancer in a xenogeneic allograft model. This tumor regression was similar to that achieved with CD28- or 4-1BB-costimulated CARs, and heightened persistence was similar to 4-1BB but greater than CD28. Thus, CD27 costimulation enhances expansion, effector function, and survival of human CAR-T cells in vitro and augments human T-cell persistence and antitumor activity in vivo.


Subject(s)
Breast Neoplasms/therapy , CD28 Antigens/immunology , Immunotherapy, Adoptive , Mesothelioma/therapy , Ovarian Neoplasms/therapy , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Animals , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/immunology , CD28 Antigens/genetics , Cell Proliferation , Cytokines/metabolism , Female , Flow Cytometry , Fluorescent Antibody Technique , Humans , Lymphocyte Activation , Mesothelioma/genetics , Mesothelioma/immunology , Mice , Mice, Inbred NOD , Mice, SCID , Ovarian Neoplasms/genetics , Ovarian Neoplasms/immunology , Receptors, Antigen, T-Cell/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
12.
Cancer Res ; 71(13): 4617-27, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21546571

ABSTRACT

Human T cells engineered to express a chimeric antigen receptor (CAR) specific for folate receptor-α (FRα) have shown robust antitumor activity against epithelial cancers in vitro but not in the clinic because of their inability to persist and home to tumor in vivo. In this study, CARs were constructed containing a FRα-specific scFv (MOv19) coupled to the T-cell receptor CD3ζ chain signaling module alone (MOv19-ζ) or in combination with the CD137 (4-1BB) costimulatory motif in tandem (MOv19-BBζ). Primary human T cells transduced to express conventional MOv19-ζ or costimulated MOv19-BBζ CARs secreted various proinflammatory cytokines, and exerted cytotoxic function when cocultured with FRα(+) tumor cells in vitro. However, only transfer of human T cells expressing the costimulated MOv19-BBζ CAR mediated tumor regression in immunodeficient mice bearing large, established FRα(+) human cancer. MOv19-BBζ CAR T-cell infusion mediated tumor regression in models of metastatic intraperitoneal, subcutaneous, and lung-involved human ovarian cancer. Importantly, tumor response was associated with the selective survival and tumor localization of human T cells in vivo and was only observed in mice receiving costimulated MOv19-BBζ CAR T cells. T-cell persistence and antitumor activity were primarily antigen-driven; however, antigen-independent CD137 signaling by CAR improved T-cell persistence but not antitumor activity in vivo. Our results show that anti-FRα CAR outfitted with CD137 costimulatory signaling in tandem overcome issues of T-cell persistence and tumor localization that limit the conventional FRα T-cell targeting strategy to provide potent antitumor activity in vivo.


Subject(s)
Folate Receptor 1/immunology , Immunotherapy, Adoptive/methods , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology , Animals , Female , Humans , Immunoglobulin Fragments/genetics , Immunoglobulin Fragments/immunology , Mice , Ovarian Neoplasms/immunology , Ovarian Neoplasms/therapy , Protein Engineering , Receptors, Antigen, T-Cell/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 9/biosynthesis , Tumor Necrosis Factor Receptor Superfamily, Member 9/genetics , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...