Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 16 de 16
1.
J Exp Med ; 221(3)2024 Mar 04.
Article En | MEDLINE | ID: mdl-38334978

An effective cancer therapy requires killing cancer cells and targeting the tumor microenvironment (TME). Searching for molecules critical for multiple cell types in the TME, we identified NR4A1 as one such molecule that can maintain the immune suppressive TME. Here, we establish NR4A1 as a valid target for cancer immunotherapy and describe a first-of-its-kind proteolysis-targeting chimera (PROTAC, named NR-V04) against NR4A1. NR-V04 degrades NR4A1 within hours in vitro and exhibits long-lasting NR4A1 degradation in tumors with an excellent safety profile. NR-V04 inhibits and frequently eradicates established tumors. At the mechanistic level, NR-V04 induces the tumor-infiltrating (TI) B cells and effector memory CD8+ T (Tem) cells and reduces monocytic myeloid-derived suppressor cells (m-MDSC), all of which are known to be clinically relevant immune cell populations in human melanomas. Overall, NR-V04-mediated NR4A1 degradation holds promise for enhancing anticancer immune responses and offers a new avenue for treating various types of cancers such as melanoma.


Melanoma , Myeloid-Derived Suppressor Cells , Humans , Cell Line, Tumor , Immunotherapy , Melanoma/pathology , Myeloid-Derived Suppressor Cells/pathology , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Tumor Microenvironment , Proteolysis Targeting Chimera
2.
Mol Cancer Ther ; 22(4): 471-484, 2023 04 03.
Article En | MEDLINE | ID: mdl-36780212

Tumor-associated macrophages (TAM) play an important role in maintaining the immunosuppressive state of the tumor microenvironment (TME). High levels of CD163+ TAMs specifically are associated with poor prognosis in many solid tumor types. Targeting TAMs may represent a key approach in development of the next generation of cancer immune therapeutics. Members of the leukocyte immunoglobulin-like receptor B (LILRB) family, including LILRB2 (ILT4), are known to transmit inhibitory signals in macrophages and other myeloid cells. Leveraging bulk and single cell RNA-sequencing datasets, as well as extensive immunophenotyping of human tumors, we found that LILRB2 is highly expressed on CD163+ CD11b+ cells in the TME and that LILRB2 expression correlates with CD163 expression across many tumor types. To target LILRB2, we have developed JTX-8064, a highly potent and selective antagonistic mAb. JTX-8064 blocks LILRB2 binding to its cognate ligands, including classical and nonclassical MHC molecules. In vitro, JTX-8064 drives the polarization of human macrophages and dendritic cells toward an immunostimulatory phenotype. As a result, human macrophages treated with a LILRB2 blocker are reprogrammed to increase the activation of autologous T cells in co-culture systems. Furthermore, JTX-8064 significantly potentiates the activity of anti-PD-1 in allogeneic mixed lymphocyte reaction. In a human tumor explant culture, pharmacodynamic activity of JTX-8064 was observed in monotherapy and in combination with anti-PD-1. Collectively, our work provides strong translational and preclinical rationale to target LILRB2 in cancer.


Neoplasms , Humans , Neoplasms/genetics , Neoplasms/metabolism , Macrophages/metabolism , Lymphocyte Activation , Coculture Techniques , T-Lymphocytes , Tumor Microenvironment , Membrane Glycoproteins/genetics , Receptors, Immunologic
3.
Oncoimmunology ; 11(1): 2141007, 2022.
Article En | MEDLINE | ID: mdl-36352891

The presence of T regulatory (Treg) cells in the tumor microenvironment is associated with poor prognosis and resistance to therapies aimed at reactivating anti-tumor immune responses. Therefore, depletion of tumor-infiltrating Tregs is a potential approach to overcome resistance to immunotherapy. However, identifying Treg-specific targets to drive such selective depletion is challenging. CCR8 has recently emerged as one of these potential targets. Here, we describe GS-1811, a novel therapeutic monoclonal antibody that specifically binds to human CCR8 and is designed to selectively deplete tumor-infiltrating Tregs. We validate previous findings showing restricted expression of CCR8 on tumor Tregs, and precisely quantify CCR8 receptor densities on tumor and normal tissue T cell subsets, demonstrating a window for selective depletion of Tregs in the tumor. Importantly, we show that GS-1811 depleting activity is limited to cells expressing CCR8 at levels comparable to tumor-infiltrating Tregs. Targeting CCR8 in mouse tumor models results in robust anti-tumor efficacy, which is dependent on Treg depleting activity, and synergizes with PD-1 inhibition to promote anti-tumor responses in PD-1 resistant models. Our data support clinical development of GS-1811 to target CCR8 in cancer and drive tumor Treg depletion in order to promote anti-tumor immunity.


Neoplasms , T-Lymphocytes, Regulatory , Mice , Animals , Humans , T-Lymphocytes, Regulatory/metabolism , Programmed Cell Death 1 Receptor , Immunotherapy/methods , Neoplasms/therapy , Tumor Microenvironment , Immunoglobulin Fc Fragments/metabolism , Receptors, CCR8/metabolism
4.
Nature ; 506(7486): 52-7, 2014 Feb 06.
Article En | MEDLINE | ID: mdl-24476824

Recent clinical trials showed that targeting of inhibitory receptors on T cells induces durable responses in a subset of cancer patients, despite advanced disease. However, the regulatory switches controlling T-cell function in immunosuppressive tumours are not well understood. Here we show that such inhibitory mechanisms can be systematically discovered in the tumour microenvironment. We devised an in vivo pooled short hairpin RNA (shRNA) screen in which shRNAs targeting negative regulators became highly enriched in murine tumours by releasing a block on T-cell proliferation upon tumour antigen recognition. Such shRNAs were identified by deep sequencing of the shRNA cassette from T cells infiltrating tumour or control tissues. One of the target genes was Ppp2r2d, a regulatory subunit of the PP2A phosphatase family. In tumours, Ppp2r2d knockdown inhibited T-cell apoptosis and enhanced T-cell proliferation as well as cytokine production. Key regulators of immune function can therefore be discovered in relevant tissue microenvironments.


Immunotherapy , Molecular Targeted Therapy , Protein Phosphatase 2/metabolism , Tumor Microenvironment/immunology , Animals , Antigens, Neoplasm/immunology , Apoptosis/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Proliferation , Cytokines/immunology , Cytokines/metabolism , Female , Gene Knockdown Techniques , High-Throughput Nucleotide Sequencing , Immunotherapy/methods , Lymphocytes, Tumor-Infiltrating/cytology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Melanoma, Experimental/immunology , Mice , Mice, Inbred C57BL , Protein Phosphatase 2/deficiency , Protein Phosphatase 2/genetics , RNA, Small Interfering/genetics , Reproducibility of Results
5.
Mol Ther Nucleic Acids ; 2: e105, 2013 Jul 09.
Article En | MEDLINE | ID: mdl-23839099

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.

6.
Mol Ther ; 21(8): 1611-20, 2013 Aug.
Article En | MEDLINE | ID: mdl-23732988

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.


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
7.
Mol Ther ; 21(3): 629-37, 2013 Mar.
Article En | MEDLINE | ID: mdl-23070117

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.


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
8.
J Immunother ; 35(2): 159-68, 2012.
Article En | MEDLINE | ID: mdl-22306904

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.


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
9.
Pediatr Blood Cancer ; 59(4): 758-61, 2012 Oct.
Article En | MEDLINE | ID: mdl-21994111

Severe chronic active Epstein-Barr virus infection (CAEBV) in T or NK cells is a rare complication of latent EBV infection. CAEBV associated T-cell lymphoproliferative disease (LPD) consists of polyclonal lesions as well as aggressive lymphomas. Here, we report such a patient. In addition, we show that this primary CAEBV associated T-cell lymphoma expresses CD70 and is sensitive to killing by CD70-specific T cells, identifying CD70 as a potential immunotherapeutic target for CAEBV-associated T-cell lymphoma.


CD27 Ligand/metabolism , Epstein-Barr Virus Infections/complications , Lymphoma, T-Cell/virology , T-Lymphocytes/immunology , Child, Preschool , Chronic Disease , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/therapy , Hematopoietic Stem Cell Transplantation , Humans , Lymphoma, T-Cell/etiology , Lymphoma, T-Cell/immunology , Male
10.
PLoS One ; 6(5): e20267, 2011.
Article En | MEDLINE | ID: mdl-21647415

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.


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
11.
Cytotherapy ; 13(8): 976-86, 2011 Sep.
Article En | MEDLINE | ID: mdl-21539497

BACKGROUND AIMS: Hematopoietic stem cell transplant (HSCT) is the treatment of choice for a proportion of patients with hematologic malignancies as well as for non-malignant diseases. However, viral infections, particularly Epstein-Barr virus (EBV), cytomegalovirus (CMV) and adenovirus (Ad), remain problematic after transplant despite the use of antiviral drugs. We have shown that cytotoxic T lymphocytes (CTL) generated against CMV-pp65, EBV and Ad antigens in a single culture are capable of controlling infections with all three viruses after HSCT. Although pp65-specific CTL have proved efficacious for the control of CMV infection, several reports highlight the importance of targeting additional CMV antigens. METHODS: To expand multivirus-specific T cells with activity against both CMV-pp65 and CMV-IE-1, peripheral blood mononuclear cells (PBMC) were transduced with the adenoviral vector (Ad5f35-IE-1-I-pp65). After 9-12 days the CTL were restimulated with autologous EBV-transformed B cells transduced with the same Ad vector. RESULTS: After 18 days in culture nine CTL lines expanded from less than 1.5 × 10(7) PBMC to a mean of 6.1 × 10(7) T cells that recognized CMV antigens pp65 [median 273 spot-forming cells (SFC), range 47-995] and IE-1 (median 154 SFC, range 11-505), the Ad antigens hexon (median 153 SFC, range 26-465) and penton (median 37 SFC, range 1-353), as well as EBV lymphoblastoid cell lines (median 55 SFC, range 9-301). Importantly, the T cells recognized at least two antigens per virus and lysed virus peptide-pulsed targets. CONCLUSIONS: CTL that target at least two antigens each of CMV, EBV and Ad should have clinical benefit with broad coverage of all three viruses and enhanced control of CMV infections compared with current protocols.


Adenoviridae/physiology , Cytomegalovirus/physiology , Hematologic Neoplasms/therapy , Hematopoietic Stem Cell Transplantation , Herpesvirus 4, Human/physiology , Postoperative Complications , T-Lymphocytes, Cytotoxic/metabolism , Virus Diseases/etiology , Cell Culture Techniques , Cell Proliferation , Cells, Cultured , Hematologic Neoplasms/pathology , Humans , Immediate-Early Proteins/genetics , Immediate-Early Proteins/immunology , Immediate-Early Proteins/metabolism , Lymphocyte Activation , Phosphoproteins/genetics , Phosphoproteins/immunology , Phosphoproteins/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , T-Cell Antigen Receptor Specificity/genetics , T-Cell Antigen Receptor Specificity/immunology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/pathology , T-Lymphocytes, Cytotoxic/transplantation , Viral Matrix Proteins/genetics , Viral Matrix Proteins/immunology , Viral Matrix Proteins/metabolism , Virus Diseases/prevention & control
12.
Blood ; 117(16): 4304-14, 2011 Apr 21.
Article En | MEDLINE | ID: mdl-21304103

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.


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
13.
Immunotherapy ; 2(5): 663-71, 2010 Sep.
Article En | MEDLINE | ID: mdl-20874650

Epstein-Barr virus-associated lymphoproliferative diseases (EBV-LPD) after hematopoietic stem cell transplantation or solid-organ transplantation remain a serious and potentially life-threatening complication. In the last decade, outcomes for EBV-LPD have significantly improved. Key to this success was the development of early detection methods, such as serial measurements of EBV-DNA load in the peripheral blood of transplant recipients. Immunotherapeutic interventions for EBV-LPD include reduction of immunosuppression, CD20 monoclonal antibodies (rituximab) as monotherapy or in conjunction with chemotherapy, and adoptive immunotherapy with EBV-specific T cells. Pre-emptive immunotherapeutic interventions can prevent the development of EBV-LPD. As monotherapy, immunotherapy is effective in inducing remissions of EBV-LPD with low-risk features. For high-risk disease, combining immunotherapy with conventional therapies has led to superior outcomes. Current challenges consist of risk stratifying patients so that patients receive the most efficacious therapy without suffering from unwanted side effects.


Antibodies, Monoclonal, Murine-Derived/therapeutic use , Epstein-Barr Virus Infections/therapy , Immunotherapy , Lymphoproliferative Disorders/therapy , T-Lymphocytes, Cytotoxic/transplantation , Animals , Combined Modality Therapy , Epstein-Barr Virus Infections/complications , Epstein-Barr Virus Infections/immunology , Humans , Immunosuppression Therapy/adverse effects , Lymphoproliferative Disorders/etiology , Lymphoproliferative Disorders/immunology , Rituximab , T-Lymphocytes, Cytotoxic/immunology , Transplantation
15.
J Immunother ; 32(7): 726-36, 2009 Sep.
Article En | MEDLINE | ID: mdl-19561536

An optimized antigen-presenting cell for tumor immunotherapy should produce a robust antigen specific cytotoxic T lymphocytes (CTL) response to tumor-associated antigens, which can persist in vivo and expand on antigen reencounter. Interleukin (IL)-21 synergizes with other gamma-chain cytokines to enhance the frequency and cytotoxicity of antigen-specific CTL. As T cells themselves may serve as effective antigen-presenting cells (T antigen-presenting cells; TAPC) and may be useful in vivo as cellular vaccines, we examined whether CD8(+) T cells genetically modified to produce IL-21 could induce immune responses to tumor associated antigen peptides in healthy human leukocyte antigen-A2(+) donors. We found that IL-21 modified TAPC enhanced both the proliferation and survival of MART-1 specific CD8(+) T cells, which were enriched by >8-fold over cultures with control nontransgenic TAPC. MART-1-specific CTL produced interferon-gamma in response to cognate peptide antigen and killed primary tumor cells expressing MART-1 in a major histocompatibility complex restricted manner. IL-21 modified TAPC similarly enhanced generation of functional CTL against melanoma antigen gp100 and the B-cell chronic lymphocytic leukemia associated RHAMM antigen. Antigen-specific CTL generated using IL-21 gene-modified TAPC had a central memory phenotype characterized by CD45RA(-), CD44(high), CD27(high), CD28(high), CD62L(high), and IL-7 receptor-alpha(high), contrasting with the terminal effector phenotype of CTL generated in the absence of IL-21. Thus, TAPC stimulation in the presences of IL-21 enhances proliferation of tumor antigen-specific T cells and favors induction of a central memory phenotype, which may improve proliferation, survival, and efficacy of T-cell based therapies for the treatment of cancer.


Antigen Presentation/immunology , CD4-Positive T-Lymphocytes/immunology , Interleukins/metabolism , T-Lymphocytes, Cytotoxic/immunology , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Apoptosis/drug effects , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Cell Proliferation , Cytotoxicity, Immunologic/immunology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gene Expression , Humans , Immunologic Memory/immunology , Immunophenotyping , Interleukins/genetics , Interleukins/pharmacology , K562 Cells , Lentivirus/genetics , Lymphoid Enhancer-Binding Factor 1/genetics , Lymphoid Enhancer-Binding Factor 1/metabolism , Reverse Transcriptase Polymerase Chain Reaction , T Cell Transcription Factor 1/genetics , T Cell Transcription Factor 1/metabolism , T-Lymphocytes, Cytotoxic/metabolism , Transfection
...