Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Science ; 359(6383): 1537-1542, 2018 Mar 30.
Article in English | MEDLINE | ID: mdl-29599246

ABSTRACT

MICA and MICB are expressed by many human cancers as a result of cellular stress, and can tag cells for elimination by cytotoxic lymphocytes through natural killer group 2D (NKG2D) receptor activation. However, tumors evade this immune recognition pathway through proteolytic shedding of MICA and MICB proteins. We rationally designed antibodies targeting the MICA α3 domain, the site of proteolytic shedding, and found that these antibodies prevented loss of cell surface MICA and MICB by human cancer cells. These antibodies inhibited tumor growth in multiple fully immunocompetent mouse models and reduced human melanoma metastases in a humanized mouse model. Antitumor immunity was mediated mainly by natural killer (NK) cells through activation of NKG2D and CD16 Fc receptors. This approach prevents the loss of important immunostimulatory ligands by human cancers and reactivates antitumor immunity.


Subject(s)
Antibodies, Blocking/therapeutic use , Antibodies, Monoclonal/therapeutic use , Histocompatibility Antigens Class I/immunology , Killer Cells, Natural/immunology , Melanoma/therapy , Animals , Antibodies, Blocking/immunology , Antibodies, Monoclonal/immunology , Histocompatibility Antigens Class I/chemistry , Humans , Immunocompetence , Ligands , Melanoma/immunology , Melanoma/pathology , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , NK Cell Lectin-Like Receptor Subfamily K/immunology , Neoplasm Metastasis , Protein Domains/immunology , Receptors, IgG/immunology
2.
Clin Cancer Res ; 17(16): 5233-8, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21700764

ABSTRACT

The interaction between the immune system and prostate cancer has been an area of research interest for several decades. The recent U.S. Food and Drug Administration approval of 2 first-in-class proof-of-concept immunotherapies (sipuleucel-T and ipilimumab) has stimulated broader interest in manipulating immunity to fight cancer. In the context of prostate cancer, the immunotherapy strategies that have garnered the most interest are the therapeutic vaccination strategies, exemplified by sipuleucel-T and PROSTVAC-VF, and immune checkpoint blockade of CTLA-4 and PD-1. Improved understanding of the immune responses generated by these strategies and development of predictive biomarkers for patient selection will guide rational combinations of these treatments and provide building blocks for future immunotherapies.


Subject(s)
Cancer Vaccines/therapeutic use , Immunotherapy/methods , Prostatic Neoplasms/immunology , Prostatic Neoplasms/therapy , Animals , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , CTLA-4 Antigen/immunology , CTLA-4 Antigen/metabolism , Clinical Trials as Topic , Disease Models, Animal , Humans , Immunotherapy/trends , Ipilimumab , Male , Mice , Models, Immunological , Programmed Cell Death 1 Receptor/immunology , Programmed Cell Death 1 Receptor/metabolism , Prostatic Neoplasms/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tissue Extracts/therapeutic use
3.
Blood ; 118(2): 348-57, 2011 Jul 14.
Article in English | MEDLINE | ID: mdl-21551230

ABSTRACT

Studying human antigen-specific memory B cells has been challenging because of low frequencies in peripheral blood, slow proliferation, and lack of antibody secretion. Therefore, most studies have relied on conversion of memory B cells into antibody-secreting cells by in vitro culture. To facilitate direct ex vivo isolation, we generated fluorescent antigen tetramers for characterization of memory B cells by using tetanus toxoid as a model antigen. Brightly labeled memory B cells were identified even 4 years after last immunization, despite low frequencies ranging from 0.01% to 0.11% of class-switched memory B cells. A direct comparison of monomeric to tetrameric antigen labeling demonstrated that a substantial fraction of the B-cell repertoire can be missed when monomeric antigens are used. The specificity of the method was confirmed by antibody reconstruction from single-cell sorted tetramer(+) B cells with single-cell RT-PCR of the B-cell receptor. All antibodies bound to tetanus antigen with high affinity, ranging from 0.23 to 2.2 nM. Furthermore, sequence analysis identified related memory B cell and plasmablast clones isolated more than a year apart. Therefore, antigen tetramers enable specific and sensitive ex vivo characterization of rare memory B cells as well as the production of fully human antibodies.


Subject(s)
Antigens/immunology , B-Lymphocytes/cytology , Cell Separation/methods , Immunologic Memory , Animals , Antibody Specificity , Antigens/metabolism , Antigens/pharmacology , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , CHO Cells , Cells, Cultured , Cricetinae , Cricetulus , Humans , Immunologic Memory/immunology , Models, Biological , Protein Multimerization/immunology , Protein Multimerization/physiology , Tetanus Toxoid/chemical synthesis , Tetanus Toxoid/immunology , Tetanus Toxoid/metabolism
4.
J Immunol ; 178(3): 1542-52, 2007 Feb 01.
Article in English | MEDLINE | ID: mdl-17237403

ABSTRACT

CD4(+)CD25(+) regulatory T cells (Tregs) suppress immunity to infections and tumors as well as autoimmunity and graft-vs-host disease. Since Tregs constitutively express CTLA-4 and activated T cells express B7-1 and B7-2, it has been suggested that the interaction between CTLA-4 on Tregs and B7-1/2 on the effector T cells may be required for immune suppression. In this study, we report that autopathogenic T cells from B7-deficient mice cause multiorgan inflammation when adoptively transferred into syngeneic RAG-1-deficient hosts. More importantly, this inflammation is suppressed by adoptive transfer of purified wild-type (WT) CD4(+)CD25(+) T cells. WT Tregs also inhibited lymphoproliferation and acquisition of activation markers by the B7-deficient T cells. An in vitro suppressor assay revealed that WT and B7-deficient T cells are equally susceptible to WT Treg regulation. These results demonstrate that B7-deficient T cells are highly susceptible to immune suppression by WT Tregs and refute the hypothesis that B7-CTLA-4 interaction between effector T cells and Tregs plays an essential role in Treg function.


Subject(s)
Antigens, CD/metabolism , Antigens, Differentiation/metabolism , B7-1 Antigen/physiology , Immune Tolerance , T-Lymphocytes, Regulatory/physiology , T-Lymphocytes/immunology , Adoptive Transfer , Animals , Autoimmunity , B7-1 Antigen/genetics , B7-1 Antigen/metabolism , CTLA-4 Antigen , Cell Proliferation , Inflammation , Lymphocyte Activation , Mice , Mice, Knockout , T-Lymphocytes/pathology , T-Lymphocytes/transplantation , T-Lymphocytes, Regulatory/transplantation
5.
Blood ; 109(1): 253-8, 2007 Jan 01.
Article in English | MEDLINE | ID: mdl-16896159

ABSTRACT

The impact of timing of antigen introduction into fetus and neonates leads to the suggestion that pre-existing antigens are tolerogenic to immunocompetent cells generated thereafter. This hypothesis predicts that in patients with cancer who are undergoing bone marrow transplantation, newly produced T cells with specificity for pre-existing tumor cells will be inactivated by the tumor antigens in the host. Because the effect of tumor cells on developing cancer-reactive T cells has not been investigated, we set out to systematically analyze the impact of tumor cells in the periphery on the development of tumor-reactive T cells in the thymus and their immunocompetence in the periphery. Our data demonstrate that in the host in which a tumor is established in the periphery, the cancer-reactive T cells develop normally, remain fully immunocompetent, become activated in the periphery, and cause regression of large established tumors. The immunocompetence of T cells generated in an antigen-bearing host is also confirmed in a skin graft transplantation model.


Subject(s)
Antigens, Neoplasm/immunology , Bone Marrow Transplantation , Immunocompetence , Plasmacytoma/immunology , T-Lymphocyte Subsets/immunology , Animals , Clonal Deletion , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Mice, Transgenic , Models, Immunological , Plasmacytoma/therapy , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Skin Neoplasms/immunology , Specific Pathogen-Free Organisms , T-Cell Antigen Receptor Specificity , Transplantation, Homologous
6.
Cancer Res ; 66(14): 7276-84, 2006 Jul 15.
Article in English | MEDLINE | ID: mdl-16849577

ABSTRACT

The majority of cancer antigens identified thus far have limited expression in normal tissues. It has been suggested that autoimmune disease is a necessary price for cancer immunity. This notion is supported by a recent clinical trial involving an anti-CTL antigen-4 (CTLA-4) antibody that showed significant clinical responses but severe autoimmune diseases in melanoma patients. To selectively modulate cancer immunity and autoimmunity, we used anti-CTLA-4 and anti-4-1BB antibodies to treat mice with a preexisting cancer, MC38. The combination of the two antibodies led to CD8 T-cell-mediated rejection of large established MC38 tumors and long-lasting immunity to the same tumor cells, although the same regimen was not effective for B16 melanoma. More importantly, whereas individual antibodies induced inflammation and autoimmune manifestations, combination therapy increased cancer immunity while reducing autoimmunity. The reduction of autoimmune effects correlates with an increased function of regulatory T cells. Our results suggest a novel approach to simultaneously enhance cancer immunity and reduce autoimmunity.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antigens, CD/immunology , Antigens, Differentiation/immunology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Colonic Neoplasms/therapy , Receptors, Nerve Growth Factor/immunology , Receptors, Tumor Necrosis Factor/immunology , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/immunology , Antineoplastic Combined Chemotherapy Protocols/immunology , Autoimmunity/immunology , CD8-Positive T-Lymphocytes/immunology , CTLA-4 Antigen , Colonic Neoplasms/immunology , Female , Humans , Immunization, Passive/methods , Mice , Mice, Inbred C57BL , T-Lymphocytes, Regulatory/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9
7.
Blood ; 106(9): 3127-33, 2005 Nov 01.
Article in English | MEDLINE | ID: mdl-16037385

ABSTRACT

Although results from preclinical studies in animal models have proven the concept for use of anti-cytotoxic T-lymphocyte antigen 4 (CTLA-4) antibodies in cancer immunotherapy, 2 major obstacles have hindered their successful application for human cancer therapy. First, the lack of in vitro correlates of the antitumor effect of the antibodies makes it difficult to screen for the most efficacious antibody by in vitro analysis. Second, significant autoimmune side effects have been observed in a recent clinical trial. In order to address these 2 issues, we have generated human CTLA4 gene knock-in mice and used them to compare a panel of anti-human CTLA-4 antibodies for their ability to induce tumor rejection and autoimmunity. Surprisingly, while all antibodies induced protection against cancer and demonstrated some autoimmune side effects, the antibody that induced the strongest protection also induced the least autoimmune side effects. These results demonstrate that autoimmune disease does not quantitatively correlate with cancer immunity. Our approach may be generally applicable to the development of human therapeutic antibodies.


Subject(s)
Antibodies/immunology , Antigens, Differentiation/immunology , Antigens, Differentiation/metabolism , Autoimmunity/immunology , Neoplasms/immunology , Animals , Antibodies/pharmacology , Antigens, CD , Antigens, Differentiation/genetics , Autoimmunity/drug effects , CTLA-4 Antigen , Cell Line , Complement C3/metabolism , Humans , Kidney/immunology , Kidney/metabolism , Mice , Mice, Transgenic , Neoplasms/genetics , Neoplasms/metabolism
8.
Blood ; 105(3): 1114-20, 2005 Feb 01.
Article in English | MEDLINE | ID: mdl-15486062

ABSTRACT

When adopting basic principles learned in mice to clinical application in humans, it is often difficult to distinguish whether a "translation" fails because of an invalid target in the human disease or because the therapeutic agents are not optimal for the human target. It is, therefore, desirable to develop preclinical models to optimize therapies for human targets using in vivo settings. Although anti-mouse CTLA-4 antibodies are known to enhance immune responses in vivo, their effect on T-cell activation in vitro ranges from enhancement to inhibition. Here we use the hu-PBL-SCID mouse model of Epstein-Barr virus (EBV)-associated lymphoma development to screen a panel of anti-human CTLA-4 monoclonal antibodies (mAbs) for their effect on human lymphocytes in an in vivo "humanized" environment. We report significant heterogeneity of anti-human CTLA-4 mAbs in enhancing the expansion of human T cells in mice, and this heterogeneity cannot be attributed to immunoglobulin isotypes or affinity for CTLA-4. These data validate the development of additional screening tools, such as the one described, to further characterize functional activity of antihuman antibodies before proceeding with clinical translation to human studies.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antigens, Differentiation/immunology , T-Lymphocytes/immunology , Animals , Antigens, CD/drug effects , Antigens, CD/immunology , CTLA-4 Antigen , Cytokines/pharmacology , Flow Cytometry , Humans , Kinetics , Leukocytes/immunology , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, SCID , Models, Animal , T-Lymphocytes/drug effects
9.
Cancer Res ; 64(21): 8062-7, 2004 Nov 01.
Article in English | MEDLINE | ID: mdl-15520217

ABSTRACT

Adoptive immunotherapy with tumor-specific T cells has emerged as a valid approach for prevention or treatment of diseases, such as melanoma and EBV-associated lymphoma. As interleukin (IL) 15 promotes survival of CD8(+) memory CTLs, we hypothesized that it could be used to enhance antitumor immunity in vivo through the maintenance of adoptively transferred memory CTL. To test this, we treated mice bearing P1A(+) tumors with adoptively transferred T cells possessing a transgenic Valpha8(+) T-cell receptor specific for the P1A tumor antigen (called P1CTL). Mice were then randomized to receive daily low-dose IL-15 (0.5 microg/day) or PBS. Mice receiving the transgenic P1CTL and IL-15 experienced a significantly delayed tumor relapse or complete tumor regression (P < 0.002 compared with PBS), with a striking persistence of the CD8(+) Valpha8(+) P1CTL compared with mice receiving the CD8(+) Valpha8(+) P1CTL and PBS vehicle (26.3 versus 5.1% P < 10(-5)). Animals exhibiting complete tumor regression had a significant population of CD8(+) Valpha8(+) P1CTL (46%) that persisted with IL-15 treatment until 140 days after adoptive transfer and successfully defended them against tumor rechallenge without IL-15. Low-dose IL-2 afforded no protection over vehicle and resulted in lower percentages of T cells with an activated memory phenotype, lower Bcl-2 expression, and lower ex vivo antitumor cytotoxicity compared with mice treated with IL-15. Collectively, the data support the notion that exogenous low-dose IL-15 therapy can enhance and even reverse the limited efficacy of adoptively transferred tumor-specific T-cell therapy and may do so in a fashion that is superior and distinct from exogenous IL-2 therapy.


Subject(s)
Immunotherapy, Adoptive , Interleukin-15/therapeutic use , Interleukin-2/therapeutic use , Neoplasms, Experimental/therapy , T-Lymphocytes/immunology , Animals , Female , Immunologic Memory , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Neoplasms, Experimental/immunology , Proto-Oncogene Proteins c-bcl-2/analysis
10.
Cancer Res ; 63(18): 6051-5, 2003 Sep 15.
Article in English | MEDLINE | ID: mdl-14522934

ABSTRACT

We investigate here whether P1A, which was the first CTL-recognized and unmutated tumor antigen to be identified, is a tumor rejection antigen for J558 plasmacytoma in mice with an unperturbed T-cell repertoire. We show that although transgenic mice expressing P1A in the thymus have almost complete deletion of P1A-reactive T cells, they reject the B7-1-transfected J558 at a rate comparable with wild-type mice. Thus, P1A is not a necessary tumor rejection antigen for the J558 tumor cells. On the other hand, if anti-P1A CTL response is sufficient for tumor rejection, tumor cells must lose the antigenic epitope to evade CTL destruction. To test this, we analyze whether tumor cells escaping J558-B7 immune spleen cells harbor mutations in the P1A epitope. We find that although the spleen contained a high proportion of P1A-reactive T cells, the recurrent tumor cells have no mutation in the P1A antigenic epitope and remain susceptible to lysis by P1CTL. Thus, the antigen-bearing tumor cells can evade immune destruction in the presence of a high number of P1A-reactive T cells. Taken together, our results demonstrate that in mice with a normal TCR repertoire, substantial numbers of P1A-reactive T cells are neither necessary nor sufficient for tumor rejection and raise interesting questions regarding the significance of T-cell response against unmutated tumor antigens.


Subject(s)
Antigens, Neoplasm/immunology , Neoplasms, Experimental/immunology , T-Lymphocytes/immunology , Animals , Antigens, Neoplasm/genetics , Epitopes, T-Lymphocyte/immunology , Immune Tolerance , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Neoplasms, Experimental/prevention & control , Transfection
11.
Cancer Res ; 62(12): 3459-65, 2002 Jun 15.
Article in English | MEDLINE | ID: mdl-12067989

ABSTRACT

Anti-4-1BB monoclonal antibody (mAb) has been shown to induce antitumor immunity by a CD4/CD8-dependent mechanism, but its direct effect on tumor-specific CD8+ T cells in tumor rejection is unclear. Here we used transgenic CD8+ T cells against the unmutated tumor rejection antigen P1A to analyze whether this mAb can promote CD8+ T-cell function against large tumors in the absence of CD4+ T-helper cells. RAG-2(-/-) mice were challenged with P1A-expressing plasmacytoma J558. Once tumor size reached a diameter of 0.85-1.75 cm, mice were treated with P1A-specific CD8+ CTL (P1CTL) in conjunction with anti-4-1BB mAb or control IgG. All of the mice showed a partial regression of tumor, but mice treated with anti-4-1BB mAb exhibited markedly enhanced tumor rejection, delayed tumor progression, and prolonged survival. Correspondingly, we observed a substantial increase in the number of P1CTL in anti-4-1BB mAb-treated mice. Surprisingly, anti-4-1BB mAb did not accelerate division of the tumor-specific CD8+ T cells, and the increase in tumor-specific T-cell number was due to reduced activation-induced cell death. These results indicate that anti-4-1BB mAb can promote CD8+ T cell-mediated protection against large tumors in the absence of CD4+ T-cell help by promoting P1CTL survival without increasing initial clonal expansion.


Subject(s)
Antibodies, Monoclonal/immunology , CD8-Positive T-Lymphocytes/immunology , Immunotherapy, Adoptive/methods , Receptors, Nerve Growth Factor/immunology , Receptors, Tumor Necrosis Factor/immunology , Animals , Antibodies, Monoclonal/pharmacology , Antigens, CD , Antigens, Neoplasm/immunology , Cell Death/immunology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , Epitopes, T-Lymphocyte/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Plasmacytoma/immunology , Plasmacytoma/therapy , T-Lymphocytes, Cytotoxic/immunology , Tumor Cells, Cultured , Tumor Necrosis Factor Receptor Superfamily, Member 9
12.
Blood ; 99(8): 2880-9, 2002 Apr 15.
Article in English | MEDLINE | ID: mdl-11929778

ABSTRACT

Costimulatory molecules B7-1 and B7-2 (hereby collectively called B7) interact with CD28 and CTLA4 on T cells and promote antitumor immunity. The function of B7-CTLA4 interaction in antitumor CTL response remains controversial. Here we used CD28(-/-) and CD28(+/-) or CD28(+/+) transgenic mice that express the T-cell receptor specific for an unmutated tumor antigen, P1A, and for tumor cells expressing a CTLA4-specific B7 mutant to evaluate the function of CD28-B7 and CTLA4-B7 interactions in induction and effector phases of antitumor immunity. We report that B7-CD28 and B7-CTLA4 interactions promote tumor rejection. However, this is achieved by distinct mechanisms. B7-CD28 interaction enhances T-cell clonal expansion, though a role for this interaction in the effector phase cannot be ruled out. In contrast, B7-CTLA4 interaction enhances the CTL-mediated destruction of tumors, but not T-cell clonal expansion.


Subject(s)
Antigens, Differentiation/metabolism , Antigens, Neoplasm/immunology , B7-1 Antigen/metabolism , Immunoconjugates , T-Lymphocytes, Cytotoxic/immunology , Abatacept , Adoptive Transfer , Animals , Antigens, CD/genetics , Antigens, CD/immunology , Antigens, CD/metabolism , Antigens, Differentiation/immunology , B7-1 Antigen/genetics , B7-1 Antigen/immunology , B7-2 Antigen , CD28 Antigens/genetics , CD28 Antigens/metabolism , CTLA-4 Antigen , Cytotoxicity, Immunologic/drug effects , Immunity , Lymphocyte Activation/drug effects , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Membrane Glycoproteins/metabolism , Mice , Mice, Transgenic , Mutation , Neoplasms, Experimental/therapy , Protein Binding/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes, Cytotoxic/transplantation
SELECTION OF CITATIONS
SEARCH DETAIL
...