551
|
Cavallo F, Offringa R, van der Burg SH, Forni G, Melief CJM. Vaccination for Treatment and Prevention of Cancer in Animal Models. Adv Immunol 2006; 90:175-213. [PMID: 16730264 DOI: 10.1016/s0065-2776(06)90005-4] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Two approaches to immunological intervention in tumor-host interactions in mouse models are discussed in this review. The first is described with reference to experiments in which CD8(+) T lymphocytes are used to kill established transplantable tumors. Peptides and their optimal presentation by dendritic cells and intervention in immune regulatory mechanisms are the key issues for efficient induction of T-killer cell-mediated tumor eradication. The time frame of tumor therapy and the threat imposed by tumor growth in transplantable models and cancer patients require the induction of a robust T-cell reaction. Prevention of the progression of small preneoplastic lesions, on the other hand, requires the significant and prolonged immune protection sought in the second approach. This is based on antibody production and the coordinated activation of multiple low-avidity cell-mediated mechanisms elicited by DNA vaccination in genetically modified cancer-prone mice, transgenic for a mutant Her-2/neu growth factor receptor expressed at the plasma membrane surface of preneoplastic mammary gland epithelial cells. Vaccination with appropriate DNA formulations results in prolonged immune inhibition of the progression of preneoplastic mammary lesions but is ineffective against established tumors. The use of molecularly defined adjuvants and intervention in immune regulatory mechanisms are critical in both the elicitation of an effective T-cell mediated reaction required for tumor debulking in the first set of models and the induction by vaccination of a sustained immune memory able to prevent the expansion of preneoplastic lesions in genetically cancer-prone mice.
Collapse
Affiliation(s)
- Federica Cavallo
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
| | | | | | | | | |
Collapse
|
552
|
Barber DL, Wherry EJ, Masopust D, Zhu B, Allison JP, Sharpe AH, Freeman GJ, Ahmed R. Restoring function in exhausted CD8 T cells during chronic viral infection. Nature 2005; 439:682-7. [PMID: 16382236 DOI: 10.1038/nature04444] [Citation(s) in RCA: 3053] [Impact Index Per Article: 160.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2005] [Accepted: 11/21/2005] [Indexed: 12/11/2022]
Abstract
Functional impairment of antigen-specific T cells is a defining characteristic of many chronic infections, but the underlying mechanisms of T-cell dysfunction are not well understood. To address this question, we analysed genes expressed in functionally impaired virus-specific CD8 T cells present in mice chronically infected with lymphocytic choriomeningitis virus (LCMV), and compared these with the gene profile of functional memory CD8 T cells. Here we report that PD-1 (programmed death 1; also known as Pdcd1) was selectively upregulated by the exhausted T cells, and that in vivo administration of antibodies that blocked the interaction of this inhibitory receptor with its ligand, PD-L1 (also known as B7-H1), enhanced T-cell responses. Notably, we found that even in persistently infected mice that were lacking CD4 T-cell help, blockade of the PD-1/PD-L1 inhibitory pathway had a beneficial effect on the 'helpless' CD8 T cells, restoring their ability to undergo proliferation, secrete cytokines, kill infected cells and decrease viral load. Blockade of the CTLA-4 (cytotoxic T-lymphocyte-associated protein 4) inhibitory pathway had no effect on either T-cell function or viral control. These studies identify a specific mechanism of T-cell exhaustion and define a potentially effective immunological strategy for the treatment of chronic viral infections.
Collapse
Affiliation(s)
- Daniel L Barber
- Emory Vaccine Center and Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | | | | | | | | | | | | | | |
Collapse
|
553
|
Abstract
The most significant recent advances in the application of monoclonal antibodies (mAbs) to oncology have been the introduction and approval of bevacizumab (Avastin), an anti-vascular endothelial growth factor antibody, and of cetuximab (Erbitux), an anti-epidermal growth factor antibody. In combination with standard chemotherapy regimens, bevacizumab significantly prolongs the survival of patients with metastatic cancers of the colorectum, breast and lung. Cetuximab, used alone or with salvage chemotherapy, produces clinically meaningful anti-tumor responses in patients with chemotherapy-refractory cancers of the colon and rectum. In addition, the anti-HER2/neu antibody trastuzumab (Herceptin), in combination with standard adjuvant chemotherapy, has been shown to reduce relapses and prolong disease-free and overall survival in high-risk patients after definitive local therapy for breast cancer. These exciting recent results provide optimism for the development of mAbs that bind novel targets, exploit novel mechanisms of action or possess improved tumor targeting. Progress in the clinical use of radioimmunoconjugates remains hindered by complexity of administration, toxicity concerns and insufficiently selective tumor targeting.
Collapse
Affiliation(s)
- Gregory P Adams
- Department of Medical Oncology, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, Pennsylvania 19111, USA
| | | |
Collapse
|
554
|
Evel-Kabler K, Song XT, Aldrich M, Huang XF, Chen SY. SOCS1 restricts dendritic cells' ability to break self tolerance and induce antitumor immunity by regulating IL-12 production and signaling. J Clin Invest 2005; 116:90-100. [PMID: 16357940 PMCID: PMC1312019 DOI: 10.1172/jci26169] [Citation(s) in RCA: 136] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2005] [Accepted: 09/18/2005] [Indexed: 12/11/2022] Open
Abstract
DC-based tumor vaccine research has largely focused on enhancing DC maturation/costimulation and antigen presentation in order to break tolerance against self tumor-associated antigens. DC immunization can activate autoreactive T cells but rarely causes autoimmune pathologies, indicating that self tolerance at the host level is still maintained in the vaccinated hosts. This study in mice reveals a novel regulatory mechanism for the control of self tolerance at the host level by DCs through the restriction of positive cytokine feedback loops by cytokine signaling inhibitor SOCS1. The study further finds the requirement of persistent antigen presentation by DCs for inducing pathological autoimmune responses against normal tissues and tumor, which can be achieved by silencing SOCS1 to unleash the unbridled signaling of IL-12 and the downstream cytokine cascade. However, the use of higher-affinity self peptides, enhancement of DC maturation, and persistent stimulation with cytokines or TLR agonists fail to break tolerance and induce pathological antitumor immunity. Thus, this study indicates the necessity of inhibiting SOCS1, an antigen presentation attenuator, to break self tolerance and induce effective antitumor responses.
Collapse
Affiliation(s)
- Kevin Evel-Kabler
- Center for Cell and Gene Therapy, Department of Immunology, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | | | | | |
Collapse
|
555
|
Childs B, Wiener C, Valle D. A science of the individual: implications for a medical school curriculum. Annu Rev Genomics Hum Genet 2005; 6:313-30. [PMID: 16124864 DOI: 10.1146/annurev.genom.6.080604.162345] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Barton Childs
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | | | | |
Collapse
|
556
|
Demaria S, Bhardwaj N, McBride WH, Formenti SC. Combining radiotherapy and immunotherapy: a revived partnership. Int J Radiat Oncol Biol Phys 2005; 63:655-66. [PMID: 16199306 PMCID: PMC1489884 DOI: 10.1016/j.ijrobp.2005.06.032] [Citation(s) in RCA: 271] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2005] [Revised: 06/28/2005] [Accepted: 06/30/2005] [Indexed: 02/07/2023]
Abstract
Ionizing radiation therapy (RT) is an important local modality for the treatment of cancer. The current rationale for its use is based largely on the ability of RT to kill the cancer cells by a direct cytotoxic effect. Nevertheless, considerable evidence indicates that RT effects extend beyond the mere elimination of the more radiosensitive fraction of cancer cells present within a tumor at the time of radiation exposure. For instance, a large body of evidence is accumulating on the ability of RT to modify the tumor microenvironment and generate inflammation. This might have far-reaching consequences regarding the response of a patient to treatment, especially if radiation-induced tumor cell kill were to translate into the generation of effective antitumor immunity. Although much remains to be learned about how radiation can impact tumor immunogenicity, data from preclinical studies provide the proof of principle that different immunotherapeutic strategies can be combined with RT to enhance antitumor effects. Conversely, RT could be a useful tool to combine with immunotherapy. This article will briefly summarize what is known about the impact of RT on tumor immunity, including tumor-associated antigens, antigen-presenting cells, and effector mechanisms. In addition, the experimental evidence supporting the contention that RT can be used as a tool to induce antitumor immunity is discussed, and a new approach to radioimmunotherapy of cancer is proposed.
Collapse
Affiliation(s)
- Sandra Demaria
- Departments of Pathology and
- Address correspondence and reprint requests to: Sandra Demaria, M. D., Department of Pathology, MSB-563, New York University School of Medicine, 550 First Avenue, New York, NY 10016. Tel: (212) 263-7308; Fax: (212) 263-8211; e-mail:
| | - Nina Bhardwaj
- NYU Cancer Institute, New York University School of Medicine, New York, NY 10016
| | - William H. McBride
- Department of Radiation Oncology, Experimental Division, University of California at Los Angeles School of Medicine, Los Angeles, CA 90095, USA
| | | |
Collapse
|
557
|
Abstract
T cell activation requires interactions of T cell antigen receptors (TCR) and peptides presented by major histocompatibility complex molecules (MHCp) in an adhesive junction between the T cell and antigen-presenting cell. Stable junctions with bull's eye supramolecular activation clusters (SMACs) have been defined as immunological synapses (IS). These structures maintain T cell-APC interaction and allow directed secretion. T cells can also be activated by asymmetric hemi-synapses (HS) that allow migration during signal integration. IS and HS operate in different stages of T cell priming. Optimal effector functions may also depend upon cyclical use of IS and HS.
Collapse
Affiliation(s)
- Michael L Dustin
- Program in Molecular Pathogenesis, Skirball Institute of Biomolecular Medicine, and Department of Pathology, New York University School of Medicine, 540 1st Ave, New York, NY 10016, USA.
| |
Collapse
|
558
|
Abstract
Lymphoproliferative disorders, including follicular lymphoma (FL), multiple myeloma (MM) and chronic lymphatic leukaemia (CLL), are slowly progressive malignancies which remain incurable despite advances in therapy. Harnessing the immune system to recognise and destroy tumours is a promising new approach to treating these diseases. Dendritic cells (DC) are unique antigen-presenting cells that play a central role in the initiation and direction of immune responses. DC loaded ex vivo with tumour-associated antigens and administered as a vaccine have already shown promise in early clinical trials for a number of lymphoproliferative disorders, but the need for improvement is widely agreed. Recent advances in the understanding of basic DC biology and lessons from early clinical trials have provided exciting new insights into the generation of anti-tumour immune responses and the design of vaccine strategies. In this review we provide an overview of our current understanding of DC biology and their function in patients with lymphoproliferative disorders. We discuss the current status of clinical trials and new approaches to exploit the antigen presenting capacity of DC to design vaccines of the future.
Collapse
MESH Headings
- Cancer Vaccines/immunology
- Cancer Vaccines/therapeutic use
- Clinical Trials as Topic
- Dendritic Cells/immunology
- Dendritic Cells/pathology
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Leukemia, Lymphocytic, Chronic, B-Cell/therapy
- Lymphoma, Follicular/immunology
- Lymphoma, Follicular/pathology
- Lymphoma, Follicular/therapy
- Lymphoproliferative Disorders/immunology
- Lymphoproliferative Disorders/pathology
- Lymphoproliferative Disorders/therapy
- Multiple Myeloma/immunology
- Multiple Myeloma/pathology
- Multiple Myeloma/therapy
- Vaccination
Collapse
Affiliation(s)
- Kristen J Radford
- Mater Medical Research Institute, Dendritic Cell Laboratory, South Brisbane, Queensland, Australia.
| | | | | |
Collapse
|
559
|
Maker AV, Phan GQ, Attia P, Yang JC, Sherry RM, Topalian SL, Kammula US, Royal RE, Haworth LR, Levy C, Kleiner D, Mavroukakis SA, Yellin M, Rosenberg SA. Tumor regression and autoimmunity in patients treated with cytotoxic T lymphocyte-associated antigen 4 blockade and interleukin 2: a phase I/II study. Ann Surg Oncol 2005; 12:1005-16. [PMID: 16283570 PMCID: PMC1473970 DOI: 10.1245/aso.2005.03.536] [Citation(s) in RCA: 377] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2005] [Accepted: 07/20/2005] [Indexed: 12/16/2022]
Abstract
BACKGROUND Cytotoxic T lymphocyte-associated antigen (CTLA)-4 can inhibit T-cell responses and is involved in tolerance against self antigens. We previously reported autoimmune manifestations and objective cancer regressions in patients with metastatic melanoma treated with CTLA-4 blockade. The possibility of activating tumor-reactive T cells while removing inhibitory activity with CTLA-4 blockade has stimulated interest in using anti-CTLA-4 antibodies in combination with other cancer immunotherapies to improve clinical outcomes. In this study, we assessed the antitumor activity and autoimmune toxicity of CTLA-4 blockade in combination with an immune-activating stimulus, interleukin (IL)-2, in patients with metastatic melanoma. METHODS Thirty-six patients received anti-CTLA-4 antibody every 3 weeks. Three patients per cohort received doses of .1, .3, 1.0, and 2.0 mg/kg. Twenty-four patients received 3.0 mg/kg. All patients received IL-2 therapy (720,000 IU/kg every 8 hours to a maximum of 15 doses). RESULTS Eight patients (22%) experienced objective tumor responses (three complete and five partial), including metastases in the lungs, lymph nodes, mediastinum, and subcutaneous tissues. Six of the eight patients have ongoing objective responses at 11 to 19 months. Five patients (14%) developed grade III/IV autoimmune toxicities secondary to anti-CTLA-4 administration, including four patients with enterocolitis and one with arthritis and uveitis. CONCLUSIONS There is not evidence to support a synergistic effect of CTLA-4 blockade plus IL-2 administration, because the 22% objective response rate is that expected from the sum of these two agents administered alone. Durable cancer regressions were seen in patients treated with this combination.
Collapse
Affiliation(s)
- Ajay V. Maker
- Surgery Branch, National Cancer Institute, National Institutes of Health, CRC Room 3–3940, 10 Center Drive, MSC 1201, Bethesda, Maryland 20814
| | - Giao Q. Phan
- Surgery Branch, National Cancer Institute, National Institutes of Health, CRC Room 3–3940, 10 Center Drive, MSC 1201, Bethesda, Maryland 20814
| | - Peter Attia
- Surgery Branch, National Cancer Institute, National Institutes of Health, CRC Room 3–3940, 10 Center Drive, MSC 1201, Bethesda, Maryland 20814
| | - James C. Yang
- Surgery Branch, National Cancer Institute, National Institutes of Health, CRC Room 3–3940, 10 Center Drive, MSC 1201, Bethesda, Maryland 20814
| | - Richard M. Sherry
- Surgery Branch, National Cancer Institute, National Institutes of Health, CRC Room 3–3940, 10 Center Drive, MSC 1201, Bethesda, Maryland 20814
| | - Suzanne L. Topalian
- Surgery Branch, National Cancer Institute, National Institutes of Health, CRC Room 3–3940, 10 Center Drive, MSC 1201, Bethesda, Maryland 20814
| | - Udai S. Kammula
- Surgery Branch, National Cancer Institute, National Institutes of Health, CRC Room 3–3940, 10 Center Drive, MSC 1201, Bethesda, Maryland 20814
| | - Richard E. Royal
- Surgery Branch, National Cancer Institute, National Institutes of Health, CRC Room 3–3940, 10 Center Drive, MSC 1201, Bethesda, Maryland 20814
| | - Leah R. Haworth
- Surgery Branch, National Cancer Institute, National Institutes of Health, CRC Room 3–3940, 10 Center Drive, MSC 1201, Bethesda, Maryland 20814
| | - Catherine Levy
- Surgery Branch, National Cancer Institute, National Institutes of Health, CRC Room 3–3940, 10 Center Drive, MSC 1201, Bethesda, Maryland 20814
| | - David Kleiner
- Surgery Branch, National Cancer Institute, National Institutes of Health, CRC Room 3–3940, 10 Center Drive, MSC 1201, Bethesda, Maryland 20814
| | - Sharon A. Mavroukakis
- Surgery Branch, National Cancer Institute, National Institutes of Health, CRC Room 3–3940, 10 Center Drive, MSC 1201, Bethesda, Maryland 20814
| | - Michael Yellin
- Medarex, Inc., 519 Route 173 West, Bloomsbury, New Jersey 08804
| | - Steven A. Rosenberg
- Surgery Branch, National Cancer Institute, National Institutes of Health, CRC Room 3–3940, 10 Center Drive, MSC 1201, Bethesda, Maryland 20814
- Address correspondence and reprint requests to: Steven A. Rosenberg, MD, PhD; E-mail:
| |
Collapse
|
560
|
Abstract
Immune function is notoriously complex, and current biomedical research elaborates this complexity by focusing on the cellular and molecular mechanisms that characterize immune defenses. However, the human immune system is a product of natural selection that develops and functions in whole organisms that are integral parts of their surrounding environments. A population-level, cross-cultural, adaptationist perspective is therefore a necessary complement to the micro levels of analysis currently favored by biomedical immunology. Prior field-based research on human immunity is reviewed to demonstrate the relevance of cultural ecological factors, with an emphasis on the ecologies of nutrition, infectious disease, reproduction, and psychosocial stress. Common themes and anthropological contributions are identified in an attempt to promote future research in human ecological immunology that integrates theory and method for a more contextualized understanding of this important physiological system.
Collapse
Affiliation(s)
- Thomas W. McDade
- Department of Anthropology, Northwestern University, Evanston, Illinois 60208
| |
Collapse
|
561
|
Sabel MS, Hess SD, Egilmez NK, Conway TF, Chen FA, Bankert RB. CTLA-4 blockade augments human T lymphocyte-mediated suppression of lung tumor xenografts in SCID mice. Cancer Immunol Immunother 2005; 54:944-52. [PMID: 15846492 PMCID: PMC11032930 DOI: 10.1007/s00262-005-0668-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2004] [Accepted: 12/29/2004] [Indexed: 11/28/2022]
Abstract
Previous studies by others using transplantable murine tumor models have demonstrated that the administration of antibodies that block CTLA-4 interaction with B7 can provoke the elimination of established tumors, and that the tumor suppression is mediated by T-cells and/or cells expressing NK1.1. Studies from our lab have established in a human/severe combined immunodeficient (SCID) mouse chimeric model that autologous peripheral blood leukocytes (PBL) can suppress the growth of tumor xenografts in a PBL dose-dependent fashion, and that this suppression is dependent upon the patient's T and NK cells. Using this human/mouse chimeric model, we sought to determine whether an antibody blockade of CTLA-4 would enhance the anti-tumor response of a patient's PBL. It was first important to determine whether the tumor suppression observed in the SCID model was dependent upon CD28/B7 co-stimulation. Blockade of B7 with a human CTLA-4-Ig fusion protein completely abrogated the lymphocyte-mediated tumor suppression, confirming in this model that tumor suppression is dependent upon a CD28/B7 co-stimulation. Using two different CTLA-4 specific monoclonal antibodies, we observed that CTLA-4 blockade significantly enhanced the human lymphocyte-mediated tumor suppression in mice co-engrafted with PBL and tumor cells. This enhancement was observed in both an allogeneic setting (in which the PBL were allogeneic with respect to the tumor) and an autologous setting (in which the PBL and tumor were from the same patient). These results sustain the notion that human anti-tumor immune response can be augmented (in vivo) by blocking the interaction between CTLA-4 and B7.
Collapse
MESH Headings
- Abatacept
- Animals
- Antibodies, Monoclonal/pharmacology
- Antigens/genetics
- Antigens/immunology
- Antigens, CD/immunology
- Antigens, Differentiation/immunology
- Antigens, Ly
- Antigens, Surface
- B7-1 Antigen/immunology
- B7-2 Antigen
- CD28 Antigens/immunology
- CTLA-4 Antigen
- Carcinoma, Squamous Cell/immunology
- Carcinoma, Squamous Cell/prevention & control
- Dendritic Cells/immunology
- Humans
- Immunoconjugates/pharmacology
- Killer Cells, Natural/immunology
- Lectins, C-Type
- Lung Neoplasms/immunology
- Lung Neoplasms/prevention & control
- Lymphocyte Activation
- Membrane Glycoproteins/immunology
- Mice
- Mice, SCID
- NK Cell Lectin-Like Receptor Subfamily B
- Proteins/genetics
- Proteins/immunology
- Severe Combined Immunodeficiency/immunology
- T-Lymphocytes/immunology
- Transplantation, Heterologous
Collapse
Affiliation(s)
- Michael S Sabel
- Department of Surgery, 3304 Cancer Center, University of Michigan, 1500 East Medical Center Drive, Ann Arbor, MI 48109, USA.
| | | | | | | | | | | |
Collapse
|
562
|
Chen YQ, Shi HZ, Qin XJ, Mo WN, Liang XD, Huang ZX, Yang HB, Wu C. CD4+CD25+ regulatory T lymphocytes in malignant pleural effusion. Am J Respir Crit Care Med 2005; 172:1434-9. [PMID: 16151041 DOI: 10.1164/rccm.200504-588oc] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Active suppression by CD4(+)CD25(+) regulatory T lymphocytes plays an important role in the downregulation of T-cell responses to foreign and self-antigens. OBJECTIVE To analyze whether the CD4(+)CD25(+) regulatory T lymphocytes exist and function normally in malignant pleural effusion. METHODS The percentages of CD4(+)CD25(+) T lymphocytes in pleural effusion and peripheral blood from patients with lung cancer with malignant effusion, pleural lavage and peripheral blood from patients with lung cancer without effusion, and peripheral blood from healthy control subjects were determined by flow cytometry. The expressions of forkhead transcription factor Foxp3 and cytotoxic lymphocyte-associated antigen-4 were also examined. CD4(+)CD25(+) and CD4(+)CD25(-) T cells from pleural effusion and peripheral blood were isolated, and were cultured to observe the effects of CD4(+)CD25(+) cells on proliferation response of CD4(+)CD25(-) T cells in vitro. MAIN RESULTS There were increased numbers of CD4(+)CD25(+) T cells in malignant pleural effusion from patients with lung cancer compared with pleural lavage from patients with lung cancer without pleural effusion, and that these cells have constitutive high-level expression of Foxp3 and cytotoxic lymphocyte-associated antigen-4. Furthermore, CD4(+)CD25(+) T cells mediate potent inhibition of proliferation response of CD4(+)CD25(-) T cells, and anticytotoxic lymphocyte-associated antigen-4 monoclonal antibody could reduce the inhibitory activity of CD4(+)CD25(+) T cells. CONCLUSIONS The increased CD4(+)CD25(+) T cells found in malignant pleural effusion express high levels of Foxp3 transcription factor and potently suppress the proliferation of CD4(+)CD25(-) T cells, and cytotoxic lymphocyte-associated antigen-4 is involved in the suppressive activity of pleural CD4(+)CD25(+) T cells.
Collapse
MESH Headings
- Adult
- Aged
- Antibodies, Monoclonal/pharmacology
- Antigens, CD
- Antigens, Differentiation/pharmacology
- CD4 Antigens/drug effects
- CD4 Antigens/genetics
- CD4 Antigens/immunology
- CTLA-4 Antigen
- Cell Proliferation
- Female
- Flow Cytometry
- Forkhead Transcription Factors/genetics
- Forkhead Transcription Factors/metabolism
- Humans
- Immunoglobulin Fc Fragments/pharmacology
- Immunosuppressive Agents/pharmacology
- Male
- Middle Aged
- Pleural Effusion, Malignant/drug therapy
- Pleural Effusion, Malignant/immunology
- Pleural Effusion, Malignant/metabolism
- RNA, Messenger/genetics
- Receptors, Interleukin-2/drug effects
- Receptors, Interleukin-2/genetics
- Receptors, Interleukin-2/immunology
- Reverse Transcriptase Polymerase Chain Reaction
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/pathology
Collapse
Affiliation(s)
- Yi-Qiang Chen
- Institute of Respiratory Diseases, First Affiliated Hospital, Guangxi Medical University, Nanning 530021, Guangxi, P. R. China.
| | | | | | | | | | | | | | | |
Collapse
|
563
|
Cheung TC, Humphreys IR, Potter KG, Norris PS, Shumway HM, Tran BR, Patterson G, Jean-Jacques R, Yoon M, Spear PG, Murphy KM, Lurain NS, Benedict CA, Ware CF. Evolutionarily divergent herpesviruses modulate T cell activation by targeting the herpesvirus entry mediator cosignaling pathway. Proc Natl Acad Sci U S A 2005; 102:13218-23. [PMID: 16131544 PMCID: PMC1201609 DOI: 10.1073/pnas.0506172102] [Citation(s) in RCA: 166] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The herpesvirus entry mediator (HVEM), a member of the TNF receptor (TNFR) superfamily, can act as a molecular switch that modulates T cell activation by propagating positive signals from the TNF-related ligand LIGHT (TNFR superfamily 14), or inhibitory signals through the Ig superfamily member B and T lymphocyte attenuator (BTLA). Competitive binding analysis and mutagenesis reveals a unique BTLA binding site centered on a critical lysine residue in cysteine-rich domain 1 of HVEM. The BTLA binding site on HVEM overlaps with the binding site for the herpes simplex virus 1 envelope glycoprotein D, but is distinct from where LIGHT binds, yet glycoprotein D inhibits the binding of both ligands, potentially nullifying the pathway. The binding site on HVEM for BTLA is conserved in the orphan TNFR, UL144, present in human CMV. UL144 binds BTLA, but not LIGHT, and inhibits T cell proliferation, selectively mimicking the inhibitory cosignaling function of HVEM. The demonstration that distinct herpesviruses target the HVEM-BTLA cosignaling pathway suggests the importance of this pathway in regulating T cell activation during host defenses.
Collapse
MESH Headings
- Base Sequence
- Binding Sites
- Binding, Competitive
- Cytomegalovirus/immunology
- Cytomegalovirus/physiology
- Herpesviridae/immunology
- Herpesviridae/physiology
- Herpesvirus 1, Human/immunology
- Herpesvirus 1, Human/physiology
- Humans
- Lymphocyte Activation/immunology
- Membrane Glycoproteins/metabolism
- Membrane Proteins/metabolism
- Molecular Sequence Data
- Phylogeny
- Receptors, Immunologic/metabolism
- Receptors, Tumor Necrosis Factor/immunology
- Receptors, Tumor Necrosis Factor/metabolism
- Receptors, Tumor Necrosis Factor/physiology
- Receptors, Tumor Necrosis Factor, Member 14
- Receptors, Virus/immunology
- Receptors, Virus/metabolism
- Receptors, Virus/physiology
- Signal Transduction
- T-Lymphocytes/immunology
- T-Lymphocytes/virology
- Tumor Necrosis Factor Ligand Superfamily Member 14
- Tumor Necrosis Factor-alpha/metabolism
- Viral Envelope Proteins/metabolism
- Viral Proteins/metabolism
Collapse
Affiliation(s)
- Timothy C Cheung
- Division of Molecular Immunology, La Jolla Institute for Allergy and Immunology, 10355 Science Center Drive, San Diego, CA 92121, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
564
|
Posnett DN, Engelhorn ME, Houghton AN. Antiviral T cell responses: phalanx or multipronged attack? ACTA ACUST UNITED AC 2005; 201:1881-4. [PMID: 15967819 PMCID: PMC2212042 DOI: 10.1084/jem.20050928] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Around 700 BCE, a new military formation called the phalanx was established in ancient Greece: a tight column of heavy infantry carrying long spears, or pikes, used in a single prong of attack. Later, in the battle of Marathon described by Herodotus, the Greeks learned the advantages of multipronged attacks, a strategy still used in modern warfare. Is the immune system similar in its approach to combating pathogens or tumors?
Collapse
Affiliation(s)
- David N Posnett
- Department of Medicine, Division of Hematology-Oncology, Weill Medical College, Cornell University, New York, NY 10021, USA.
| | | | | |
Collapse
|
565
|
Attia P, Phan GQ, Maker AV, Robinson MR, Quezado MM, Yang JC, Sherry RM, Topalian SL, Kammula US, Royal RE, Restifo NP, Haworth LR, Levy C, Mavroukakis SA, Nichol G, Yellin MJ, Rosenberg SA. Autoimmunity correlates with tumor regression in patients with metastatic melanoma treated with anti-cytotoxic T-lymphocyte antigen-4. J Clin Oncol 2005; 23:6043-53. [PMID: 16087944 PMCID: PMC1473965 DOI: 10.1200/jco.2005.06.205] [Citation(s) in RCA: 761] [Impact Index Per Article: 40.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
PURPOSE Previously, we reported our experience treating 14 patients with metastatic melanoma using a fully human antibody to cytotoxic T-lymphocyte antigen-4 (anti-CTLA-4) in conjunction with peptide vaccination. We have now treated 56 patients to evaluate two different dose schedules of anti-CTLA-4 and to explore the relationship between autoimmunity and tumor regression. PATIENTS AND METHODS A total of 56 patients with progressive stage IV melanoma were enrolled onto the study. All had Karnofsky performance status > or = 60% with no prior history of autoimmunity. Twenty-nine patients received 3 mg/kg anti-CTLA-4 every 3 weeks, whereas 27 received 3 mg/kg as their initial dose with subsequent doses reduced to 1 mg/kg every 3 weeks. In both cohorts patients received concomitant vaccination with two modified HLA-A*0201-restricted peptides from the gp100 melanoma-associated antigen, gp100:209-217(210M) and gp100:280-288(288V). RESULTS Two patients achieved a complete response (ongoing at 30 and 31 months, respectively) and five patients achieved a partial response (durations of 4, 6, 25+, 26+, and 34+ months, respectively), for an overall objective response rate of 13%. Tumor regression was seen in lung, liver, brain, lymph nodes, and subcutaneous sites. Of 14 patients with grade 3/4 autoimmune toxicity, five (36%) experienced a clinical response compared with only two responses in the 42 patients (5%) with no autoimmune toxicity (P = .008). There were no significant differences in response rate or toxicity between the two dose schedules. CONCLUSION Administration of anti-CTLA-4 monoclonal antibody plus peptide vaccination can cause durable objective responses, which correlate with the induction of autoimmunity, in patients with metastatic melanoma.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Steven A. Rosenberg
- Address reprint requests to Steven A. Rosenberg, MD, PhD, Surgery Branch, National Cancer Institute, National Institutes of Health, CRC, Room 3W-3940, 10 Center Dr, Bethesda, MD 20892-1201; e-mail:
| |
Collapse
|
566
|
Boudewijns M, Jeurissen A, Wuyts M, Moens L, Boon L, Van Neerven JJ, Kasran A, Overbergh L, Lenaerts C, Waer M, Mathieu C, Ceuppens JL, Bossuyt X. Blockade of CTLA-4 (CD152) enhances the murine antibody response to pneumococcal capsular polysaccharides. J Leukoc Biol 2005; 78:1060-9. [PMID: 16081596 DOI: 10.1189/jlb.1004562] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The capsular polysaccharides (caps-PS) of Streptococcus pneumoniae are classified as thymus-independent antigens. Nevertheless, T lymphocytes can modulate the antibody response to caps-PS. In this study, we show that anticytotoxic T lymphocyte-associated antigen 4 (CTLA-4) treatment, along with administration of caps-PS to BALB/c mice, resulted in a dose-dependent generation of a strong caps-PS-specific antibody response. Anti-CTLA-4 treatment had no effect on the immunoglobulin G (IgG) antibody production in athymic nu/nu mice. Anti-CTLA-4 treatment stimulated the IgG antibody production in severe combined immunodeficiency (SCID)/SCID mice reconstituted with CTLA-4(-/-) B lymphocytes and wild-type T lymphocytes. This excluded the possibility that anti-CTLA-4 enhanced antibody production by direct interaction with B lymphocytes. Anti-CTLA-4 treatment enhanced the antibody production in SCID/SCID mice reconstituted with B lymphocytes and CD4(+) and CD8(+) T lymphocytes but not in SCID/SCID mice reconstituted with B lymphocytes in the absence of CD4(+) and/or CD8(+) cells. Administration of anti-CTLA-4 in BALB/c mice but not in nu/nu mice resulted in a markedly increased production of interleukin (IL)-2, IL-4, and interferon-gamma. Taken together, these data strongly suggest a role of T lymphocytes and CTLA-4 in the regulation of the antibody response to caps-PS.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Antibodies, Blocking/immunology
- Antibodies, Blocking/pharmacology
- Antigens, CD
- Antigens, Differentiation/drug effects
- Antigens, Differentiation/immunology
- B-Lymphocytes/immunology
- B-Lymphocytes/transplantation
- CTLA-4 Antigen
- Cytokines/genetics
- Cytokines/immunology
- Dose-Response Relationship, Drug
- Immunization
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Mice, SCID
- Pneumococcal Infections/immunology
- Pneumococcal Infections/prevention & control
- Pneumococcal Vaccines/immunology
- Pneumococcal Vaccines/pharmacology
- Polysaccharides, Bacterial/drug effects
- Polysaccharides, Bacterial/immunology
- RNA, Messenger/genetics
- RNA, Messenger/immunology
- Streptococcus pneumoniae/drug effects
- Streptococcus pneumoniae/immunology
- Survival Rate
- T-Lymphocytes/immunology
- T-Lymphocytes/transplantation
Collapse
Affiliation(s)
- Michaël Boudewijns
- Experimental Laboratory Medicine, Department of Molecular Cell Biology, Faculty of Medicine, Catholic University Leuven, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
567
|
Ji Q, Gondek D, Hurwitz AA. Provision of Granulocyte-Macrophage Colony-Stimulating Factor Converts an Autoimmune Response to a Self-Antigen into an Antitumor Response. THE JOURNAL OF IMMUNOLOGY 2005; 175:1456-63. [PMID: 16034082 DOI: 10.4049/jimmunol.175.3.1456] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Many tumor Ags recognized by T cells are self-Ags. Because high avidity, self-reactive T cells are deleted in the thymus, any residual self-reactive T cells existing in the periphery are likely to be low avidity and nonresponsive due to peripheral tolerance mechanisms. Activation of these residual T cells is critical for targeting tumors for immunotherapy. In this study, we studied immune responses against the murine B16 melanoma using a tyrosinase-related protein 2 (TRP-2) peptide as a model tumor/self-Ag. Our results showed that TRP-2 peptide vaccination alone elicited a weak T cell response and modestly decreased B16 lung tumor nodules. The combination of peptide vaccination and treatment with an Ab directed against the inhibitory receptor CTLA-4 enhanced the immune response against TRP-2 peptide, inducing autoimmune depigmentation and further decreasing lung tumor nodules. However, both vaccination methods failed to protect against orthotopic (s.c.) B16 tumor challenge. The addition of an irradiated GM-CSF-expressing, amelanotic tumor cell vaccine significantly delayed s.c. B16 tumor growth. Subsequent studies revealed that provision of GM-CSF increased dendritic cell numbers in lymph nodes and spleen. Furthermore, addition of CTLA-4 blockade increased the frequency of TRP-2-specific, IFN-secreting T cells in spleen and lymph nodes. Overall, our results indicate that combining enhancement of Ag presentation with removal of CTLA-4-mediated inhibition can convert a "weaker" autoimmune response into a more potent antitumor immune response.
Collapse
MESH Headings
- Animals
- Antibodies, Blocking/administration & dosage
- Antibodies, Blocking/therapeutic use
- Antigens, CD
- Antigens, Differentiation/immunology
- Antigens, Neoplasm/administration & dosage
- Antigens, Neoplasm/immunology
- Antineoplastic Combined Chemotherapy Protocols/immunology
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Autoantigens/immunology
- CD8-Positive T-Lymphocytes/enzymology
- CD8-Positive T-Lymphocytes/immunology
- CTLA-4 Antigen
- Cancer Vaccines/administration & dosage
- Cell Line, Tumor
- Dendritic Cells/enzymology
- Dendritic Cells/immunology
- Epitopes, T-Lymphocyte/immunology
- Female
- Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage
- Granulocyte-Macrophage Colony-Stimulating Factor/physiology
- Hypopigmentation/enzymology
- Hypopigmentation/immunology
- Injections, Subcutaneous
- Intramolecular Oxidoreductases/administration & dosage
- Intramolecular Oxidoreductases/immunology
- Lung Neoplasms/immunology
- Lung Neoplasms/prevention & control
- Lung Neoplasms/secondary
- Melanoma, Experimental/enzymology
- Melanoma, Experimental/immunology
- Melanoma, Experimental/pathology
- Melanoma, Experimental/prevention & control
- Membrane Proteins/immunology
- Mice
- Mice, Inbred C57BL
- Neoplasm Transplantation
- Peptide Fragments/administration & dosage
- Peptide Fragments/immunology
Collapse
Affiliation(s)
- Qingyong Ji
- Tumor Immunity and Tolerance Section, Laboratory of Molecular Immunoregulation, National Cancer Institute, Frederick Cancer Research and Development Center, Frederick, MD 21702, USA
| | | | | |
Collapse
|
568
|
Maçon-Lemaître L, Triebel F. The negative regulatory function of the lymphocyte-activation gene-3 co-receptor (CD223) on human T cells. Immunology 2005; 115:170-8. [PMID: 15885122 PMCID: PMC1782137 DOI: 10.1111/j.1365-2567.2005.02145.x] [Citation(s) in RCA: 144] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Accumulating evidence indicates that the CD4 homologue lymphocyte activation gene-3 (LAG-3) plays a down-regulatory role on T-cell responses. However, the role of LAG-3/major histocompatibility complex (MHC) class II interactions on primary human T-cell responses, as well as the mechanism by which down-regulation occurs, are not clear. Here, we show that LAG-3 colocalized with CD3, CD4 or CD8 in areas of cholesterol-rich raft aggregation during this primary response, as well as in the clustered raft region formed between T cells and antibody-coated beads. Addition of a blocking LAG-3-specific monoclonal antibody to both CD4 and CD8 primary resting T cells activated under conditions of antigen-presenting cell-driven stimulation and low antigen concentrations augments CD69 activation antigen expression, T-cell expansion and T helper 1 (Th1, but not Th2) cytokine production. Blocking LAG-3/MHC class II interactions leads to an increase in the number of cells entering division at these low concentrations of antigen and to more rounds of divisions with an accumulation of cells in the S-phase of the cell cycle. These results indicate that LAG-3 signalling inhibits early events in primary activation of human CD4 and CD8 T cells and further support a role for LAG-3 signalling in regulating the expansion of activated effector or memory T cells, either directly or indirectly through Treg suppressor activity.
Collapse
|
569
|
Abstract
The role of antibodies as therapeutic cancer vaccines includes two distinct approaches, which are summarised in this review, namely anti-idiotypic vaccines and antigen-antibody complex therapies. Bispecific antibodies directed against T cells or antigen-presenting cells are also referenced. The report focuses on theoretical issues, laboratory data on the mechanism of action, examples of humoral and cellular immune induction, and novel therapeutic advances in vaccine development. The biology of antigen processing and recent advances in the field of dendritic cell biology are critical to understanding the potent immune response induction. Future directions include combination therapies to manipulate immune regulatory mechanisms and to enhance clinical effects. Additional applications of antibodies targeting costimulatory or regulatory receptors on antigen-presenting cells and T cells, neutralising immune suppressive cytokines, and depleting T regulatory cells hold promise for future mono- and particularly combination therapies.
Collapse
MESH Headings
- Animals
- Antibodies/therapeutic use
- Antibodies, Anti-Idiotypic/immunology
- Antibodies, Anti-Idiotypic/therapeutic use
- Antibodies, Bispecific/immunology
- Antibodies, Bispecific/therapeutic use
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Neoplasm/biosynthesis
- Antibodies, Neoplasm/immunology
- Antigen-Antibody Complex/immunology
- Antigen-Presenting Cells/immunology
- Antigens, Neoplasm/immunology
- Cancer Vaccines/therapeutic use
- Clinical Trials as Topic
- Cytokines/antagonists & inhibitors
- Cytokines/immunology
- Dendritic Cells/immunology
- Growth Substances/immunology
- Humans
- Immunotherapy/methods
- Immunotherapy, Active
- Lymphocyte Activation
- Neoplasm Proteins/antagonists & inhibitors
- Neoplasm Proteins/immunology
- Neoplasms/immunology
- Neoplasms/therapy
- T-Lymphocytes/immunology
Collapse
Affiliation(s)
- Birgit C Schultes
- Unither Pharmaceuticals, 15 Walnut Street, Suite 300, Wellesley, MA 02481, USA.
| | | |
Collapse
|
570
|
Abstract
Blockade of antigen nonspecific costimulatory signals is a promising approach for the treatment of autoimmune diseases including systemic lupus erythematosus (SLE). CTLA4Ig, an antagonist of the CD28/B7 costimulatory interaction, effectively prevents SLE onset in several murine models and, when used in combination with cyclophosphamide, can induce remission of active SLE nephritis. In this review we describe the known mechanisms of action of CTLA4Ig both in normal immunity and in autoimmune disease models and address issues about its activity that still need to be resolved. We discuss the preclinical use of CTLA4Ig in murine SLE models and the rationale for a clinical trial in SLE patients.
Collapse
Affiliation(s)
- A Davidson
- Department of Medicine, Columbia University, New York, NY, USA.
| | | | | | | |
Collapse
|
571
|
Hodge JW, Chakraborty M, Kudo-Saito C, Garnett CT, Schlom J. Multiple costimulatory modalities enhance CTL avidity. THE JOURNAL OF IMMUNOLOGY 2005; 174:5994-6004. [PMID: 15879092 PMCID: PMC1924685 DOI: 10.4049/jimmunol.174.10.5994] [Citation(s) in RCA: 116] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Recent studies in both animal models and clinical trials have demonstrated that the avidity of T cells is a major determinant of antitumor and antiviral immunity. In this study, we evaluated several different vaccine strategies for their ability to enhance both the quantity and avidity of CTL responses. CD8(+) T cell quantity was measured by tetramer binding precursor frequency, and avidity was measured by both tetramer dissociation and quantitative cytolytic function. We have evaluated a peptide, a viral vector expressing the Ag transgene alone, with one costimulatory molecule (B7-1), and with three costimulatory molecules (B7-1, ICAM-1, and LFA-3), with anti-CTLA-4 mAb, with GM-CSF, and combinations of the above. We have evaluated these strategies in both a foreign Ag model using beta-galactosidase as immunogen, and in a "self" Ag model, using carcinoembryonic Ag as immunogen in carcinoembryonic Ag transgenic mice. The combined use of several of these strategies was shown to enhance not only the quantity, but, to a greater magnitude, the avidity of T cells generated; a combination strategy is also shown to enhance antitumor effects. The results reported in this study thus demonstrate multiple strategies that can be used in both antitumor and antiviral vaccine settings to generate higher avidity host T cell responses.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Adjuvants, Immunologic/genetics
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/immunology
- Antigens, CD
- Antigens, Differentiation/immunology
- CTLA-4 Antigen
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/genetics
- Cancer Vaccines/immunology
- Carcinoembryonic Antigen/administration & dosage
- Carcinoembryonic Antigen/genetics
- Carcinoembryonic Antigen/immunology
- Cell Line, Tumor
- Colonic Neoplasms/immunology
- Colonic Neoplasms/pathology
- Colonic Neoplasms/prevention & control
- Combined Modality Therapy
- Cytotoxicity Tests, Immunologic/methods
- Female
- Genetic Vectors
- Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage
- Lymphocyte Activation/genetics
- Lymphocyte Activation/immunology
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Peptide Fragments/administration & dosage
- Peptide Fragments/immunology
- Peptide Fragments/metabolism
- Protein Binding/immunology
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
- beta-Galactosidase/administration & dosage
- beta-Galactosidase/genetics
- beta-Galactosidase/immunology
Collapse
Affiliation(s)
- James W Hodge
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | |
Collapse
|
572
|
Cohen GB, Kaur A, Johnson RP. Isolation of viable antigen-specific CD4 T cells by CD40L surface trapping. J Immunol Methods 2005; 302:103-15. [PMID: 15993419 DOI: 10.1016/j.jim.2005.05.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2004] [Revised: 05/04/2005] [Accepted: 05/04/2005] [Indexed: 11/24/2022]
Abstract
A number of techniques have recently been developed for the identification of antigen-specific cells, yet the ability of these techniques to identify all subclasses of memory T cells has often been overlooked. Here we describe a novel approach for the isolation of live antigen-specific CD4 T cells using CD40L and CD69 surface staining and demonstrate its utility for isolating antigen-specific rhesus macaque CD4 T cells. Critical to the success of the technique was staining for CD40L concurrent with antigen stimulation. Isolation of CD4 T cells based on CD40L/CD69 surface marker upregulation identified both effector and central memory CD4 T cells. In contrast, the majority of central memory CD4 T cells did not secrete TNFalpha or IFNgamma and thus would not be identified by techniques based on their secretion. The methodology described here therefore complements existing approaches for isolating viable antigen-specific CD4 T cells, opens new avenues for investigating human diseases in nonhuman primate animal models and may prove beneficial in instances where the induced response is largely T cell central memory restricted.
Collapse
Affiliation(s)
- George B Cohen
- Department of Biochemistry and Volen Center for Complex Systems, Brandeis University, 415 South Street, Waltham, MA 02454, USA.
| | | | | |
Collapse
|
573
|
Abstract
The discovery of new functions for the original B7 family members, together with the identification of additional B7 and CD28 family members, have revealed new ways in which the B7:CD28 family regulates T cell activation and tolerance. B7-1/B7-2:CD28 interactions not only promote initial T cell activation but also regulate self-tolerance by supporting CD4+CD25+ T regulatory cell homeostasis. CTLA-4 can exert its inhibitory effects in both B7-1/B7-2 dependent and independent fashions. B7-1 and B7-2 can signal bidirectionally by engaging CD28 and CTLA-4 on T cells and by delivering signals into B7-expressing cells. The five new B7 family members, ICOS ligand, PD-L1 (B7-H1), PD-L2 (B7-DC), B7-H3, and B7-H4 (B7x/B7-S1) are expressed on professional antigen-presenting cells as well as on cells within nonlymphoid organs, providing new means for regulating T cell activation and tolerance in peripheral tissues. The new CD28 families members, ICOS, PD-1, and BTLA, are inducibly expressed on T cells, and they have important roles in regulating previously activated T cells. PD-1 and BTLA also are expressed on B cells and may have broader immunoregulatory functions. The ICOS:ICOSL pathway appears to be particularly important for stimulating effector T cell responses and T cell-dependent B cell responses, but it also has an important role in regulating T cell tolerance. In addition, the PD-1:PD-L1/PD-L2 pathway plays a critical role in regulating T cell activation and tolerance. In this review, we revisit the roles of the B7:CD28 family members in regulating immune responses, and we discuss their therapeutic potential.
Collapse
Affiliation(s)
- Rebecca J Greenwald
- Department of Pathology, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts 02115, USA.
| | | | | |
Collapse
|
574
|
Loser K, Scherer A, Krummen MBW, Varga G, Higuchi T, Schwarz T, Sharpe AH, Grabbe S, Bluestone JA, Beissert S. An important role of CD80/CD86-CTLA-4 signaling during photocarcinogenesis in mice. THE JOURNAL OF IMMUNOLOGY 2005; 174:5298-305. [PMID: 15843526 DOI: 10.4049/jimmunol.174.9.5298] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Although previous studies have shown that altered B7 costimulation plays a critical role in UV irradiation-induced regulation of immunity, the individual roles of the B7 receptors (CD28 and CTLA-4) or the B7 family members (CD80 and CD86) have not been explored. Thus, we investigated CTLA-4 signaling during photocarcinogenesis of chronically UV-B-exposed mice using an antagonistic anti-CTLA-4 Ab. Anti-CTLA-4-treated mice developed significantly fewer UV-induced tumors. Moreover, anti-CTLA-4 treatment induced long-lasting protective immunity because progressively growing UV tumors inoculated into anti-CTLA-4- and UV-treated mice that had not developed tumors were rejected. Next, we used mice deficient for CD80, CD86, or both in photocarcinogenesis studies to assess the relative contributions of these CTLA-4 ligands. Double-deficient mice showed significantly reduced UV-induced skin tumor development, whereas CD86(-/-) mice produced skin cancer earlier compared with CD80(-/-) and control mice. The growth of UV-induced tumors appears to be controlled by UV-induced suppressor T cells, because CD80(-/-)/CD86(-/-) mice had strongly reduced numbers of UV-induced CD4(+)CD25(+) suppressor T cells. In vitro, CTLA-4 blockade inhibited the suppressor activity of UV-induced CD4(+)CD25(+) T cells, suggesting that reduced photocarcinogenesis might be due to decreased numbers or function of suppressor T cells. Together, these data indicate that blocking CD80/86-CTLA-4 signaling induced immune protection against the development of UV-induced skin tumors. Furthermore, CD86-mediated costimulation appears to play a more critical role in the protection against photocarcinogenesis than CD80.
Collapse
MESH Headings
- Animals
- Antibodies, Blocking/administration & dosage
- Antibodies, Blocking/therapeutic use
- Antigens, CD/genetics
- Antigens, CD/physiology
- Antigens, Differentiation/immunology
- Antigens, Differentiation/physiology
- B7-1 Antigen/genetics
- B7-1 Antigen/physiology
- B7-2 Antigen
- Bone Marrow Cells/immunology
- CTLA-4 Antigen
- Cell Line, Tumor
- Dendritic Cells/immunology
- Graft Rejection/genetics
- Graft Rejection/immunology
- Growth Inhibitors/administration & dosage
- Growth Inhibitors/therapeutic use
- Immunosuppressive Agents/administration & dosage
- Immunosuppressive Agents/therapeutic use
- Lymphocyte Activation/genetics
- Membrane Glycoproteins/deficiency
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/physiology
- Mice
- Mice, Inbred BALB C
- Mice, Knockout
- Neoplasm Transplantation
- Neoplasms, Radiation-Induced/genetics
- Neoplasms, Radiation-Induced/immunology
- Neoplasms, Radiation-Induced/prevention & control
- Signal Transduction/genetics
- Signal Transduction/immunology
- Skin Neoplasms/genetics
- Skin Neoplasms/immunology
- Skin Neoplasms/prevention & control
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/pathology
- T-Lymphocytes, Regulatory/radiation effects
- Ultraviolet Rays
Collapse
Affiliation(s)
- Karin Loser
- Department of Dermatology, University of Munster, Munster, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
575
|
Myers L, Croft M, Kwon BS, Mittler RS, Vella AT. Peptide-specific CD8 T regulatory cells use IFN-gamma to elaborate TGF-beta-based suppression. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2005; 174:7625-32. [PMID: 15944263 DOI: 10.4049/jimmunol.174.12.7625] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
We identified a murine peptide-specific CD8 T regulatory cell population able to suppress responding CD4 T cells. Immunization with OVA, poly(I:C), and anti-4-1BB generated a population of SIINFEKL-specific CD8 T regulatory cells that profoundly inhibited peptide-responding CD4 T cells from cellular division. The mechanism of suppression required IFN-gamma, but IFN-gamma alone was not sufficient to suppress the responding CD4 T cells. The data show that CD8 T regulatory cells were unable to suppress unless they engaged IFN-gamma. Furthermore, even in the absence of recall with peptide, the CD8 T regulatory cells suppressed CD4 responses as long as IFN-gamma was present. To examine the effector mechanism of suppression, we showed that neutralizing TGF-beta inhibited suppression because inclusion of anti-TGF-beta rescued the proliferative capacity of the responding cells. TGF-beta-based suppression was dependent completely upon the CD8 T regulatory cells being capable of binding IFN-gamma. This was the case, although peptide recall of primed IFN-gamma (-/-) or IFN-gammaR(-/-) CD8 T cells up-regulated pro-TGF-beta protein as measured by surface latency-associated peptide expression but yet were unable to suppress. Finally, we asked whether the CD8 T regulatory cells were exposed to active TGF-beta in vivo and showed that only wild-type CD8 T regulatory cells expressed the TGF-beta-dependent biomarker CD103, suggesting that latency-associated peptide expression is not always congruent with elaboration of active TGF-beta. These data define a novel mechanism whereby IFN-gamma directly stimulates CD8 T regulatory cells to elaborate TGF-beta-based suppression. Ultimately, this mechanism may permit regulation of pathogenic Th1 responses by CD8 T regulatory cells.
Collapse
MESH Headings
- Animals
- Antigens, CD/biosynthesis
- CD4-Positive T-Lymphocytes/immunology
- Cell Proliferation
- Cells, Cultured
- Egg Proteins/administration & dosage
- Egg Proteins/immunology
- Epitopes, T-Lymphocyte/administration & dosage
- Epitopes, T-Lymphocyte/immunology
- Integrin alpha Chains/biosynthesis
- Interferon-gamma/metabolism
- Interferon-gamma/physiology
- Ligands
- Lymphocyte Activation/immunology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Mutant Strains
- Mice, Transgenic
- Ovalbumin/administration & dosage
- Ovalbumin/immunology
- Peptide Fragments
- Protein Binding/immunology
- Receptors, Interferon/metabolism
- Receptors, Interferon/physiology
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Transforming Growth Factor beta/physiology
- Interferon gamma Receptor
Collapse
Affiliation(s)
- Lara Myers
- Division of Immunology, University of Connecticut Health Center, Farmington, CT 06032, USA
| | | | | | | | | |
Collapse
|
576
|
Esplugues E, Vega-Ramos J, Cartoixà D, Vazquez BN, Salaet I, Engel P, Lauzurica P. Induction of tumor NK-cell immunity by anti-CD69 antibody therapy. Blood 2005; 105:4399-406. [PMID: 15692061 DOI: 10.1182/blood-2004-10-3854] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Abstract
The leukocyte activation marker CD69 is a novel regulator of the immune response, modulating the production of cytokines including transforming growth factor-β (TGF-β). We have generated an antimurine CD69 monoclonal antibody (mAb), CD69.2.2, which down-regulates CD69 expression in vivo but does not deplete CD69-expressing cells. Therapeutic administration of CD69.2.2 to wild-type mice induces significant natural killer (NK) cell–dependent antitumor responses to major histocompatibility complex (MHC) class I low RMA-S lymphomas and to RM-1 prostatic carcinoma lung metastases. These in vivo antitumor responses are comparable to those seen in CD69-/- mice. Enhanced host NK cytotoxic activity correlates with a reduction in NK-cell TGF-β production and is independent of tumor priming. In vitro studies demonstrate the novel ability of anti-CD69 mAbs to activate resting NK cells in an Fc receptor–independent manner, resulting in a substantial increase in both NK-cell cytolytic activity and interferon γ (IFNγ) production. Modulation of the innate immune system with monoclonal antibodies to host CD69 thus provides a novel means to antagonize tumor growth and metastasis.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/therapeutic use
- Antigens, CD/immunology
- Antigens, Differentiation, T-Lymphocyte/immunology
- Cytotoxicity, Immunologic
- Immunity/drug effects
- Immunotherapy/methods
- Interferon-gamma/biosynthesis
- Killer Cells, Natural/drug effects
- Killer Cells, Natural/immunology
- Lectins, C-Type
- Lung Neoplasms/drug therapy
- Lung Neoplasms/immunology
- Lung Neoplasms/secondary
- Lymphocyte Activation/drug effects
- Lymphoma/drug therapy
- Lymphoma/immunology
- Male
- Mice
- Mice, Inbred C57BL
- Neoplasms/immunology
- Neoplasms/therapy
- Prostatic Neoplasms/drug therapy
- Prostatic Neoplasms/pathology
Collapse
Affiliation(s)
- Enric Esplugues
- Departamento de Fisiología, Universidad de Barcelona, Av Diagonal, 645 Barcelona 08028, Spain
| | | | | | | | | | | | | |
Collapse
|
577
|
González-Neira A, Calafell F, Navarro A, Lao O, Cann H, Comas D, Bertranpetit J. Geographic stratification of linkage disequilibrium: a worldwide population study in a region of chromosome 22. Hum Genomics 2005; 1:399-409. [PMID: 15606995 PMCID: PMC3500194 DOI: 10.1186/1479-7364-1-6-399] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Recent studies of haplotype diversity in a number of genomic regions have suggested that long stretches of DNA are preserved in the same chromosome, with little evidence of recombination events. The knowledge of the extent and strength of these haplotypes could become a powerful tool for future genetic analysis of complex traits. Different patterns of linkage disequilibrium (LD) have been found when comparing individuals of African and European descent, but there is scarce knowledge about the worldwide population stratification. Thus, the study of haplotype composition and the pattern of LD from a global perspective are relevant for elucidating their geographical stratification, as it may have implications in the future analysis of complex traits. We have typed 12 single nucleotide polymorphisms in a chromosome 22 region--previously described as having high LD levels in European populations -- in 39 different world populations. Haplotype structure has a clear continental structure with marked heterogeneity within some continents (Africa, America). The pattern of LD among neighbouring markers exhibits a strong clustering of all East Asian populations on the one hand and of Western Eurasian populations (including Europe) on the other, revealing only two major LD patterns, but with some very specific outliers due to specific demographic histories. Moreover, it should be taken into account that African populations are highly heterogeneous. The present results support the existence of a wide (but not total) communality in LD patterns in human populations from different continental regions, despite differences in their demographic histories, as population factors seem to be less relevant compared with genomic forces in shaping the patterns of LD.
Collapse
Affiliation(s)
- Anna González-Neira
- Unitat de Biologia Evolutiva, Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, Catalonia, Spain
| | - Francesc Calafell
- Unitat de Biologia Evolutiva, Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, Catalonia, Spain
| | - Arcadi Navarro
- Unitat de Biologia Evolutiva, Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, Catalonia, Spain
| | - Oscar Lao
- Unitat de Biologia Evolutiva, Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, Catalonia, Spain
| | - Howard Cann
- Fondation Jean-Dausset, Centre d'Étude du Polymorphisme Humain (CEPH), Paris, France
| | - David Comas
- Unitat de Biologia Evolutiva, Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, Catalonia, Spain
| | - Jaume Bertranpetit
- Unitat de Biologia Evolutiva, Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, Catalonia, Spain
| |
Collapse
|
578
|
|
579
|
Li-Sucholeiki XC, Tomita-Mitchell A, Arnold K, Glassner BJ, Thompson T, Murthy JV, Berk L, Lange C, Leong-Morgenthaler PM, MacDougall D, Munro J, Cannon D, Mistry T, Miller A, Deka C, Karger B, Gillespie KM, Ekstrøm PO, Todd JA, Thilly WG. Detection and frequency estimation of rare variants in pools of genomic DNA from large populations using mutational spectrometry. Mutat Res 2005; 570:267-80. [PMID: 15708585 DOI: 10.1016/j.mrfmmm.2004.11.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2004] [Accepted: 11/29/2004] [Indexed: 11/21/2022]
Abstract
DNA variants underlying the inheritance of risk for common diseases are expected to have a wide range of population allele frequencies. The detection and scoring of the rare alleles (at frequencies of <0.01) presents significant practical problems, including the requirement for large sample sizes and the limitations inherent in current methodologies for allele discrimination. In the present report, we have applied mutational spectrometry based on constant denaturing capillary electrophoresis (CDCE) to DNA pools from large populations in order to improve the prospects of testing the role of rare variants in common diseases on a large scale. We conducted a pilot study of the cytotoxic T lymphocyte-associated antigen-4 gene (CTLA4) in type 1 diabetes (T1D). A total of 1228 bp, comprising 98% of the CTLA4 coding sequence, all adjacent intronic mRNA splice sites, and a 3' UTR sequence were scanned for unknown point mutations in pools of genomic DNA from a control population of 10,464 young American adults and two T1D populations, one American (1799 individuals) and one from the United Kingdom (2102 individuals). The data suggest that it is unlikely that rare variants in the scanned regions of CTLA4 represent a significant proportion of T1D risk and illustrate that CDCE-based mutational spectrometry of DNA pools offers a feasible and cost-effective means of testing the role of rare variants in susceptibility to common diseases.
Collapse
|
580
|
Abstract
It has recently been shown that small interfering RNA (siRNA)-mediated inhibition of suppressors of cytokine signalling 1 (SOCS1) function enhances the immunostimulatory capacity of dendritic cells (DCs), which leads to more effective cancer vaccination. This recent study demonstrates the importance of negative regulation of DC maturation and functions for DC-based immunotherapy.
Collapse
Affiliation(s)
- Takashi Kobayashi
- Molecular and Cellular Immunology Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | | |
Collapse
|
581
|
Takahashi S, Kataoka H, Hara S, Yokosuka T, Takase K, Yamasaki S, Kobayashi W, Saito Y, Saito T. In vivo overexpression of CTLA-4 suppresses lymphoproliferative diseases and thymic negative selection. Eur J Immunol 2005; 35:399-407. [PMID: 15668914 DOI: 10.1002/eji.200324746] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Cytotoxic T lymphocyte antigen-4 (CTLA-4) induces major inhibitory signals for T cell activation. From analyses of TCR-transgenic (Tg) CTLA-4-deficient mice, it has been believed that CTLA-4 does not affect thymocyte development. To focus upon the in vivo function of CTLA-4 in thymocyte development from a different aspect, we have established Tg mice expressing either full-length CTLA-4 (FL-Tg) or a mutant CTLA-4 lacking the cytoplasmic region (truncated, TR-Tg), and analyzed thymocyte development. TR-T cells express much higher CTLA-4 on the cell surface than FL-T cells, in which most CTLA-4 was localized in intracellular vesicles. While CTLA-4-/- mice exhibit lymphoproliferative disease, neither of the Tg mice with CTLA-4-/- background developed the disorder. Although the development of thymocytes appeared normal in both Tg mice, in vivo depletion of double-positive thymocytes by injection of anti-CD3 Ab as well as the elimination of minor lymphocyte-stimulating antigen-reactive thymocytes were impaired in FL-Tg mice but not in TR-Tg mice. Functionally, cross-linking of CTLA-4 on thymocytes from FL-Tg mice, but not from TR-Tg mice, inhibited proliferation. These results reveal a potential role of CTLA-4, through its cytoplasmic domain, in the negative selection of thymocytes and in the prevention of lymphoproliferative disease.
Collapse
Affiliation(s)
- Shigekazu Takahashi
- Department of Molecular Genetics, Graduate School of Medicine, Chiba University, Chiba, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
582
|
Abstract
"Nature has provided, in the white corpuscles as you call them-in the phagocytes as we call them-a natural means of devouring and destroying all disease germs. There is at bottom only one genuinely scientific treatment for all diseases, and that is to stimulate the phagocytes." So opined B.B. in G.B. Shaw's The Doctor's Dilemma in a dramatic restatement of a key portion of Ilya Metchnikoff's Nobel Prize address: "Whenever the organism enjoys immunity, the introduction of infectious microbes is followed by the accumulation of mobile cells, of white corpuscles of the blood in particular which absorb the microbes and destroy them. The white corpuscles and the other cells capable of doing this have been designated 'phagocytes,' (i.e., devouring cells) and the whole function that ensures immunity has been given the name of 'phagocytosis'". Based on these insights into the foundation of resistance to infectious disease, Metchnikoff was awarded the 1908 Nobel Prize in Physiology or Medicine together with Paul Ehrlich (Fig. 1). Although both were cited for discoveries in immunity, the contributions of the two men seem worlds apart. Ehrlich's studies did not deal with generic responses to infection, but rather with the highly specific nature of antibodies and their relationship to the cells producing them: "As the cell receptor is obviously preformed, and the artificially produced antitoxin only the consequence, i.e. secondary, one can hardly fail to assume that the antitoxin is nothing else but discharged components of the cell, namely receptors discharged in excess". But biological systems are just that-systems-and the parts need to work together. And so we arrive, a century later, at an appreciation for just how intimately related these two seemingly disparate aspects of host defense really are.
Collapse
Affiliation(s)
- Ronald N Germain
- Lymphocyte Biology Section, Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA.
| |
Collapse
|
583
|
Hurchla MA, Sedy JR, Gavrieli M, Gavrielli M, Drake CG, Murphy TL, Murphy KM. B and T Lymphocyte Attenuator Exhibits Structural and Expression Polymorphisms and Is Highly Induced in Anergic CD4+T Cells. THE JOURNAL OF IMMUNOLOGY 2005; 174:3377-85. [PMID: 15749870 DOI: 10.4049/jimmunol.174.6.3377] [Citation(s) in RCA: 136] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
B and T lymphocyte attenuator (BTLA) was initially identified as expressed on Th1 cells and B cells, but recently reported to be expressed by macrophages, dendritic cells, and NK cells as well. To address this discrepancy we generated a panel of BTLA-specific mAbs and characterized BTLA expression under various activation conditions. We report the existence of three distinct BTLA alleles among 23 murine strains, differing both in Ig domain structure and cellular distribution of expression on lymphoid subsets. The BALB/c and MRL/lpr alleles differ at one amino acid residue, but C57BL/6 has nine additional differences and alters the predicted cysteine bonding pattern. The BALB/c BTLA allele is also expressed by B cells, T cells, and dendritic cells, but not macrophages or NK cells. However, C57BL/6 BTLA is expressed on CD11b+ macrophages and NK cells. Finally, in CD4+ T cells, BTLA is expressed most highly following Ag-specific induction of anergy in vivo, and unlike programmed death-1 and CTLA-4, not expressed by CD25+ regulatory T cells. These results clarify discrepancies regarding BTLA expression, suggest that structural and expression polymorphisms be considered when analyzing BTLA in various murine backgrounds, and indicate a possible role in anergic CD4+ T cells.
Collapse
MESH Headings
- Alleles
- Amino Acid Sequence
- Animals
- Antibodies, Monoclonal
- B-Lymphocytes/immunology
- Base Sequence
- CD4-Positive T-Lymphocytes/immunology
- Clonal Anergy
- Cricetinae
- DNA, Complementary/genetics
- Gene Expression
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Inbred MRL lpr
- Mice, Knockout
- Molecular Sequence Data
- Molecular Structure
- Polymorphism, Genetic
- Receptors, Immunologic/biosynthesis
- Receptors, Immunologic/chemistry
- Receptors, Immunologic/deficiency
- Receptors, Immunologic/genetics
- Sequence Homology, Amino Acid
- Species Specificity
Collapse
Affiliation(s)
- Michelle A Hurchla
- Department of Pathology, Center for Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | | | | | | | |
Collapse
|
584
|
Workman CJ, Vignali DAA. Negative regulation of T cell homeostasis by lymphocyte activation gene-3 (CD223). THE JOURNAL OF IMMUNOLOGY 2005; 174:688-95. [PMID: 15634887 DOI: 10.4049/jimmunol.174.2.688] [Citation(s) in RCA: 236] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Lymphocyte homeostasis is a central biological process that is tightly regulated. However, its molecular and cellular control is poorly understood. We show that aged mice deficient in lymphocyte activation gene 3 (LAG-3), an MHC class II binding CD4 homologue, have twice as many T cells as wild-type controls. CD4(+) and CD8(+) LAG-3-deficient T cells showed enhanced homeostatic expansion in lymphopenic hosts, which was abrogated by ectopic expression of wild-type LAG-3, but not by a signaling-defective mutant. In addition, in vivo treatment with anti-LAG-3 mAb resulted in enhanced T cell expansion to a level comparable to that in LAG-3-deficient cells. This deregulation of T cell homeostasis also resulted in the expansion of multiple cell types, including B cells, macrophages, granulocytes, and dendritic cells. Lastly, regulatory T cells were dependent on LAG-3 for their optimal control of T cell homeostasis. Our data suggest that LAG-3 negatively regulates T cell homeostasis by regulatory T cell-dependent and independent mechanisms.
Collapse
MESH Headings
- Aging/genetics
- Aging/immunology
- Amino Acid Motifs
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antigens, CD/genetics
- Antigens, CD/immunology
- Antigens, CD/physiology
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- CD4-Positive T-Lymphocytes/transplantation
- Cell Differentiation/immunology
- Cytoplasm/immunology
- Down-Regulation/immunology
- Homeostasis/genetics
- Homeostasis/immunology
- Injections, Intravenous
- Lymphocyte Activation/genetics
- Lymphocyte Count
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Protein Structure, Tertiary
- Receptors, Interleukin-2/biosynthesis
- T-Lymphocyte Subsets/cytology
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- T-Lymphocyte Subsets/transplantation
- Lymphocyte Activation Gene 3 Protein
Collapse
Affiliation(s)
- Creg J Workman
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | | |
Collapse
|
585
|
Abstract
The etiology of autoimmune thyroid diseases (AITD) is based on genetic and nongenetic factors. Genome-wide screening and linkage analyses have identified several chromosomal regions that are linked to AITD. These are HT-1 (on chromosome 13q33) and HT-2 (chromosome 12q22) for Hashimoto's thyroiditis (HT), and GD-1 (chromosome 14q31), GD-2 (chromosome 20q11.2), and GD-3 (chromosome Xq21) for Graves' disease (GD). Several genes have been proposed as susceptibility or immunoregulatory genes. Most promising genes are those of the major histocompatibility complex (MHC) complex (chromosome 6), the cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4) gene (chromosome 2), the CD40 (chromosome 20), the thyroglobulin gene (chromosome 8), and the autoimmune regulator gene (chromosome 21). This review summarizes evidence for pathogenetic involvement of several of these genes in various forms of autoimmune thyropathies. Most genetic data refer to GD, whereas less data are available for HT and thyroid-associated ophthalmopathy. Scarce data refer to AITD within the autoimmune polyglandular syndromes I and II. The realization of family studies in large samples from different populations might provide further insight in the genetic contribution to AITD. Data are also needed on the interaction among susceptibility genes. Finally, additional functional studies are warranted to clarify the possible role of allelic variants in the underlying pathogenic mechanisms of AITD.
Collapse
Affiliation(s)
- Manuela Dittmar
- Departments of Biology and Medicine I, Gutenberg University, Mainz 55101, Germany
| | | |
Collapse
|
586
|
Couturier JP, Root-Bernstein RS. HIV may produce inhibitory microRNAs (miRNAs) that block production of CD28, CD4 and some interleukins. J Theor Biol 2005; 235:169-84. [PMID: 15862587 DOI: 10.1016/j.jtbi.2005.01.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2004] [Revised: 10/26/2004] [Accepted: 01/04/2005] [Indexed: 11/24/2022]
Abstract
It is well-known that HIV-1 infection results in a gradual decline of the CD4+ T-lymphocytes, but the underlying mechanism of this decline is not completely understood. Research has shown that HIV-1 infection of CD4+ T cells results in decreased CD28 expression, but the mechanism of this repression is unknown. There is also substantial evidence demonstrating regulatory involvement of microRNA (miRNA) during protein expression in plants and some animals, and reports have recently been published confirming the existence of viral-encoded miRNAs. Based on these findings, we hypothesize that viral-encoded miRNA from HIV-1 may directly alter T cell, macrophage and dendritic cell activity. To investigate a potential correlation between the genomic complementarity of HIV-1 and host cell protein expression, a local alignment search was performed to assess for regions of complementarity between the HIV-1 proviral genome and the mRNA coding sequence of various proteins expressed by CD+ T cells and macrophages. Regions of complementarity with strong correlations to the currently established criteria for miRNA:target mRNA activity were found between HIV-1 and CD28, CTLA-4 and some interleukins, suggesting that HIV-1 may produce translational repression in host cells.
Collapse
|
587
|
Abstract
Educating the immune system to distinguish between self and non-self is critical to ensure that an immune response is mounted against foreign antigens and not against self. A breakdown in these mechanisms can lead to the onset of autoimmune disease. Clinical and molecular data suggest that shared immunogenetic mechanisms lead to the autoimmune process. The most studied genes and molecules are the human leukocyte antigen (HLA) region and the cytotoxic T-lymphocyte-associated 4 molecule (CTLA-4). Recently progress has been achieved in narrowing down the primary variants within both gene regions, but further work is needed to determine the function and extent of the aetiological variant(s) present. Recent exciting results also suggest a role for the newly discovered lymphoid-specific phosphatase (LYP) protein. As well as these general mechanisms, disease-specific mechanisms are beginning to be elucidated, for example the role of autoimmune regulatory element 1 (AIRE1) in autoimmune polyendocrinopathy-candidiasis ectodermal dystrophy (APECED). Taken together, these data suggest that both general and disease-specific mechanisms lead to the clinical outcome of autoimmune disease and that increased understanding of these mechanisms will improve our knowledge of how autoimmune disease occurs, eventually leading to the development of novel therapeutic agents.
Collapse
Affiliation(s)
- M J Simmonds
- Division of Medical Sciences, University of Birmingham, Institute of Biomedical Research, Birmingham B15 2TT, UK
| | | |
Collapse
|
588
|
van den Berg HA, Rand DA. Foreignness as a matter of degree: the relative immunogenicity of peptide/MHC ligands. J Theor Biol 2005; 231:535-48. [PMID: 15488530 DOI: 10.1016/j.jtbi.2004.07.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2004] [Revised: 07/07/2004] [Accepted: 07/12/2004] [Indexed: 12/31/2022]
Abstract
The ability of T lymphocytes (T cells) to recognize and attack foreign invaders while leaving healthy cells unharmed is often analysed as a discrete self/non-self dichotomy, with each peptide/MHC ligand classified as either self or non-self. We argue that the ligand immunogenicity is more naturally treated as a continuous quantity, and show how to define and quantitate relative ligand immunogenicity. In our theory, self-tolerance is acquired through reduction of the relative immunogenicity of autoantigens, whereas xenoantigens, typically not presented during induction of deletional tolerance, retain a high degree of relative immunogenicity. Autoantigens that are not prominently presented in deletional tolerance likewise retain a high relative immunogenicity and remain essentially foreign. According to our analysis, any given autoantigen can attain a high level of relative immunogenicity, provided it is presented at sufficiently high levels. Our theory provides a quantitative tool to analyse the immunogenicity of tumour-associated neoantigens and the aetiology of autoimmune disease.
Collapse
Affiliation(s)
- Hugo A van den Berg
- Interdisciplinary Programme for Cellular Regulation, Mathematics Institute, University of Warwick, Coventry CV4 7AL, UK.
| | | |
Collapse
|
589
|
Baccala R, Gonzalez-Quintial R, Dummer W, Theofilopoulos AN. Tumor immunity via homeostatic T cell proliferation: mechanistic aspects and clinical perspectives. ACTA ACUST UNITED AC 2005; 27:75-85. [PMID: 15666151 DOI: 10.1007/s00281-004-0196-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2004] [Accepted: 12/15/2004] [Indexed: 10/25/2022]
Abstract
Efforts to develop effective anti-tumor immunotherapies are hampered by the difficulty of overcoming tolerance against tumor antigens, which in most instances are normal gene products that are over-expressed, preferentially expressed or re-expressed in cancer cells. Considering that lymphopenia-induced homeostatic T cell proliferation is mediated by self-peptide/MHC recognition and that the expanded cells acquire some effector functions, we hypothesized that this process could be used to break tolerance against tumor antigens. Studies by us and others in several mouse models demonstrated that availability of tumor antigens during homeostatic T cell proliferation indeed leads to effective anti-tumor autoimmunity with specificity and memory. This effect appears to be mediated by reduction in the activation threshold of low-affinity tumor-specific T cells, leading to their preferential engagement and expansion. In its simplicity, this approach is likely to have application in humans, since it relies on conventional lymphopenia-inducing cancer therapies, infusion of autologous lymphocytes and, optimally, tumor-specific vaccination.
Collapse
Affiliation(s)
- Roberto Baccala
- Department of Immunology, IMM-3, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA.
| | | | | | | |
Collapse
|
590
|
Demaria S, Kawashima N, Yang AM, Devitt ML, Babb JS, Allison JP, Formenti SC. Immune-Mediated Inhibition of Metastases after Treatment with Local Radiation and CTLA-4 Blockade in a Mouse Model of Breast Cancer. Clin Cancer Res 2005. [DOI: 10.1158/1078-0432.728.11.2] [Citation(s) in RCA: 285] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Abstract
Purpose: Ionizing radiation therapy (RT) is an important component in the management of breast cancer. Although the primary tumor can be successfully treated by surgery and RT, metastatic breast cancer remains a therapeutic challenge. Here we tested the hypothesis that the combination of RT to the primary tumor with CTLA-4 blockade can elicit antitumor immunity inhibiting the metastases.
Experimental Design: The poorly immunogenic metastatic mouse mammary carcinoma 4T1 was used as a model. Mice were injected s.c. with 4T1 cells, and treatment was started 13 days later when the primary tumors measured 5 mm in average diameter. Mice were randomly assigned to four treatment groups receiving: (1) control IgG (IgG), (2) RT + IgG, (3) 9H10 monoclonal antibody against CTLA-4, (4) RT + 9H10. RT was delivered to the primary tumor by one or two fractions of 12 Gy. 9H10 and IgG were given i.p. thrice after RT.
Results: Consistent with the fact that 4T1 is poorly immunogenic, 9H10 alone did not have any effect on primary tumor growth or survival. RT was able to delay the growth of the primary irradiated tumor, but in the absence of 9H10 survival was similar to that of control mice. In contrast, mice treated with RT + 9H10 had a statistically significant survival advantage. The increased survival correlated with inhibition of lung metastases formation and required CD8+ but not CD4+ T cells.
Conclusions: The combination of local RT with CTLA-4 blockade is a promising new immunotherapeutic strategy against poorly immunogenic metastatic cancers.
Collapse
Affiliation(s)
| | | | | | | | - James S. Babb
- 3Radiology, New York University School of Medicine, New York, New York; and
| | - James P. Allison
- 4Howard Hughes Medical Institute, University of California at Berkeley, Berkeley, California
| | | |
Collapse
|
591
|
Kwon H, Jun HS, Khil LY, Yoon JW. Role of CTLA-4 in the activation of single- and double-positive thymocytes. THE JOURNAL OF IMMUNOLOGY 2005; 173:6645-53. [PMID: 15557155 DOI: 10.4049/jimmunol.173.11.6645] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
CTLA-4, a homologue of CD28, is a negative regulator of T cell activation in the periphery and is transiently expressed on the cell surface after T cell activation. However, the role of CTLA-4 in T cell activation in the thymus is not clear. This investigation was initiated to determine the role of CTLA-4 in the activation of CD4(+)CD8(+) double-positive (DP) and CD4(+)CD8(-) and CD4(-)CD8(+) single-positive (SP) thymocytes using fetal thymic organ cultures (FTOC) of MHC class II-restricted, OVA(323-339)-restricted TCR transgenic mice (DO11.10). We found that treatment of the FTOC with anti-CTLA-4-blocking Ab during activation with OVA(323-339) increased the proportion and number of DP thymocytes, but decreased the proportion and number of SP thymocytes compared with OVA(323-339)-stimulated FTOC without anti-CTLA-4 Ab treatment. In addition, anti-CTLA-4 Ab treatment inhibited OVA(323-339)-induced expression of the early activation marker, CD69, in DP thymocytes, but increased CD69 in SP thymocytes. Similarly, CTLA-4 blockage decreased phosphorylation of ERK in DP thymocytes by Ag-specific TCR engagement, but increased phosphorylation of ERK in SP thymocytes. CTLA-4 blockage inhibited deletion of DP thymocytes treated with a high dose of OVA(323-339), whereas CTLA-4 blockage did not inhibit deletion of DP thymocytes treated with a low dose of OVA(323-339). We conclude that CTLA-4 positively regulates the activation of DP thymocytes, resulting in their deletion, whereas blocking CTLA-4 suppresses the activation of DP thymocytes, leading to inhibition of DP thymocyte deletion. In contrast, CTLA-4 negatively regulates the activation of SP thymocytes.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antibodies, Blocking/pharmacology
- Antigens, CD
- Antigens, Differentiation/biosynthesis
- Antigens, Differentiation/genetics
- Antigens, Differentiation/immunology
- Antigens, Differentiation/physiology
- CD4 Antigens/biosynthesis
- CD8 Antigens/biosynthesis
- CTLA-4 Antigen
- Clonal Deletion
- Down-Regulation/immunology
- Epitopes, T-Lymphocyte/immunology
- Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors
- Extracellular Signal-Regulated MAP Kinases/metabolism
- Female
- Lymphocyte Activation/immunology
- Lymphocyte Count
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Molecular Sequence Data
- Organ Culture Techniques
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Signal Transduction/immunology
- T-Lymphocyte Subsets/enzymology
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- Thymus Gland/cytology
- Thymus Gland/enzymology
- Thymus Gland/immunology
- Thymus Gland/metabolism
- Up-Regulation/immunology
Collapse
Affiliation(s)
- Hyokjoon Kwon
- Julia McFarlane Diabetes Research Center, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
| | | | | | | |
Collapse
|
592
|
Abstract
The ability of activating immune recognition receptors on lymphocytes to regulate cellular activation and function can be profoundly altered by co-stimulation with inhibitory receptors. Inhibitory receptors, such as the MHC-recognizing inhibitory receptors expressed on NK cells and subpopulations of activated T cells, can fully block the generation of any cytotoxic function by targeting proximal signals. Inhibitory Fc receptors on B cells, macrophages and mast cells can influence their threshold for activation, but the induction of inhibitory Fc receptors also appears to play a major role in the attenuation of ongoing responses. The three identified groups of inhibitory B7-recognizing receptors (CTLA-4, PD-1 and BTLA) are only expressed on activated hematopoietic cells, thus exclusively regulating ongoing immune responses in lymphoid organs and the periphery. In each case, the integrated positive and negative regulatory events determine the nature of the functional response.
Collapse
Affiliation(s)
- Paul J Leibson
- Department of Immunology, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN 55905, USA.
| |
Collapse
|
593
|
Taneja V, Taneja N, Behrens M, Griffiths MM, Luthra HS, David CS. Requirement for CD28 May Not Be Absolute for Collagen-Induced Arthritis: Study with HLA-DQ8 Transgenic Mice. THE JOURNAL OF IMMUNOLOGY 2005; 174:1118-25. [PMID: 15634938 DOI: 10.4049/jimmunol.174.2.1118] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
CD28 is required to achieve optimal T cell activation to an Ag. To determine the role CD28 costimulation plays in collagen-induced arthritis, we have generated DQ8 transgenic, CD28-deficient mice. DQ8 mice deficient for CD28 had comparable numbers of CD4 and CD8 T cells as DQ8.CD28(+/+) mice. DQ8.CD28(-/-) mice develop collagen-induced arthritis with delayed onset and less severity than DQ8.CD28(+/+) mice. T cells from DQ8.CD28(-/-) mice did not respond to type II collagen efficiently in vitro, although the response to DQ8-restricted peptides was similar to that in the parent mice. There was no functional defect in T cells as observed by proliferation with Con A. Cytokine analysis from in vitro study showed the production of high levels of the inflammatory cytokine, IFN-gamma, in response to type II collagen. We observed an increase in CD4(+)CD28(-)NKG2D(+) cells after immunization, suggesting an important role for cells bearing this receptor in the disease process. CD28(-/-) mice also have an increased number of DX5(+) cells compared with CD28(+/+) mice, which can lead to the production of high levels of IFN-gamma. DQ8.CD28(-/-) mice had an increased number of cells bearing other costimulatory markers. Cells from DQ8.CD28(-/-) mice exhibited a lower proliferation rate and were resistant to activation-induced cell death compared with DQ8.CD28(+/+) mice. This study supports the idea that CD28 plays a crucial role in the regulation of arthritis. However, in the absence of CD28 signaling, other costimulatory molecules can lead to the development of disease, thus indicating that the requirement for CD28 may not be absolute in the development of arthritis.
Collapse
Affiliation(s)
- Veena Taneja
- Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA.
| | | | | | | | | | | |
Collapse
|
594
|
Abstract
Gene expression profiling provides a quantitative molecular framework for the study of human lymphomas. This genomic technology has revealed that existing diagnostic categories are comprised of multiple molecularly and clinically distinct diseases. Diffuse large B-cell lymphoma (DLBCL), for example, consists of three gene expression subgroups, termed germinal center B-cell-like (GCB) DLBCL, activated B-cell-like (ABC) DLBCL, and primary mediastinal B-cell lymphoma (PMBL). These DLBCL subgroups arise from different stages of normal B-cell differentiation, utilize distinct oncogenic mechanisms, and differ in their ability to be cured by chemotherapy. Key regulatory factors and their target genes are differentially expressed among these subgroups, including BCL-6, Blimp-1, and XBP1. ABC DLBCL and PMBL depend upon constitutive activation of the NF-kappaB pathway for their survival but GCB DLBCL does not, demonstrating that this pathway is a potential therapeutic target for certain DLBCL subgroups. In DLBCL, mantle cell lymphoma, and follicular lymphoma, gene expression profiling has also been used to create gene expression-based models of survival, which have identified the biological characteristics of the tumors that influence their clinical behavior. In mantle cell lymphoma, the length of survival following diagnosis is primarily influenced by the tumor proliferation rate, which can be quantitatively measured by a proliferation gene expression "signature." Based on this accurate measure, the proliferation rate can now be viewed as an integration of several oncogenic lesions that each increase progression from the G1 to the S phase of the cell cycle. In DLBCL and follicular lymphoma, gene expression profiling has revealed that the molecular characteristics of non-malignant tumor-infiltrating immune cells have a major influence on the length of survival. The implications of these insights for the diagnosis and treatment of non-Hodgkin lymphomas are discussed.
Collapse
MESH Headings
- B-Lymphocytes/immunology
- B-Lymphocytes/pathology
- Cell Differentiation
- Gene Expression Profiling
- Genes, MHC Class II
- Germinal Center/pathology
- Hodgkin Disease/genetics
- Humans
- Lymphoma/classification
- Lymphoma/genetics
- Lymphoma/immunology
- Lymphoma, B-Cell/classification
- Lymphoma, B-Cell/genetics
- Lymphoma, B-Cell/immunology
- Lymphoma, Follicular/genetics
- Lymphoma, Large B-Cell, Diffuse/classification
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/immunology
- Lymphoma, Mantle-Cell/genetics
- Mediastinal Neoplasms/classification
- Mediastinal Neoplasms/genetics
- Mediastinal Neoplasms/immunology
- Oncogenes
- Prognosis
Collapse
Affiliation(s)
- Louis M Staudt
- Metabolism Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland 20892, USA
| | | |
Collapse
|
595
|
Lindley S, Dayan CM, Bishop A, Roep BO, Peakman M, Tree TIM. Defective suppressor function in CD4(+)CD25(+) T-cells from patients with type 1 diabetes. Diabetes 2005; 54:92-9. [PMID: 15616015 DOI: 10.2337/diabetes.54.1.92] [Citation(s) in RCA: 634] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Type 1 diabetes is a T-cell-mediated disease that is associated with loss of immunological tolerance to self-antigens. The mechanisms involved in maintenance of peripheral tolerance include a specialized subset of regulatory T-cells (Treg) within the CD4(+)CD25(+) T-cell population, but the function and phenotype of these cells in type 1 diabetes have not been investigated. We hypothesized that a deficiency in the CD4(+)CD25(+) Treg population or its function could contribute to the lack of self-tolerance evident in patients with type 1 diabetes. We show that although levels of CD4(+)CD25(+) T-cells are normal in patients with recent-onset adult type 1 diabetes, the ability of the Tregs in this population to suppress T-cell proliferation during in vitro cocultures is markedly reduced compared with control subjects (P = 0.007). Moreover, in patients with type 1 diabetes, these cocultures display a more proinflammatory phenotype, with increased secretion of interferon-gamma (P = 0.005) and decreased interleukin-10 production (P = 0.03). These deficiencies may reflect a disturbance in the balance of the CD4(+)CD25(+) population, because in patients with type 1 diabetes, a higher proportion of these cells coexpress the early activation marker CD69 (P = 0.007) and intracellular CTLA-4 (P = 0.01). These data demonstrate deficiency in function of the CD4(+)CD25(+) Treg population that may influence the pathogenesis of type 1 diabetes.
Collapse
Affiliation(s)
- Shelley Lindley
- Department of Immunobiology, Guy's, King's and St Thomas' School of Medicine, 2nd Floor, New Guy's House, Guy's Hospital London SE1 9RT, UK
| | | | | | | | | | | |
Collapse
|
596
|
Park JM, Terabe M, van den Broeke LT, Donaldson DD, Berzofsky JA. Unmasking immunosurveillance against a syngeneic colon cancer by elimination of CD4+ NKT regulatory cells and IL-13. Int J Cancer 2005; 114:80-7. [PMID: 15523692 DOI: 10.1002/ijc.20669] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
We have previously observed a novel role of natural killer T (NKT) cells in negative regulation of antitumor immune responses against an immunogenic regressor tumor expressing a transfected viral antigen. Here, we investigated whether hidden spontaneous antitumor immunosurveillance, in the absence of a vaccine, could be revealed by disruption of this negative regulatory pathway involving CD4+ NKT cells and interleukin-13 (IL-13), in a murine pulmonary metastasis model of a nontransfected, nonregressor, syngeneic tumor, the CT26 colon carcinoma. Lung metastases of CT26 were decreased in CD4+ T cell-depleted BALB/c mice, suggesting that CD4+ T cells were involved in negative regulation of antitumor responses. CD1-knock out (CD1-KO) mice, which have conventional CD4+ T cells and CD4+CD25+ regulatory T cells but lack CD1-restricted CD4+ NKT cells, were significantly resistant to lung metastasis of CT26. The metastases were not further decreased in CD4+ T cell-depleted CD1-KO mice, implying that CD4+ NKT cells might be the primary negative regulator of antitumor immune responses in BALB/c mice. CD8+ T cells were found to act as effectors in antitumor immune responses, since the inhibition of lung metastases observed in naive CD1-KO or CD4+ T cell-depleted mice was abrogated by depletion of CD8+ T cells. Lung metastases were significantly decreased by treatment of mice with an IL-13 inhibitor, but not by deficiency or inhibition of IL-4. Thus, even for a nonregressor tumor, immunosurveillance exists but is negatively regulated via CD4+ NKT cells possibly mediated by IL-13, and can be unmasked by removal of these negative regulatory components.
Collapse
Affiliation(s)
- Jong Myun Park
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | |
Collapse
|
597
|
Inobe M, Schwartz RH. CTLA-4 Engagement Acts as a Brake on CD4+ T Cell Proliferation and Cytokine Production but Is Not Required for Tuning T Cell Reactivity in Adaptive Tolerance. THE JOURNAL OF IMMUNOLOGY 2004; 173:7239-48. [PMID: 15585846 DOI: 10.4049/jimmunol.173.12.7239] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Adaptive tolerance is the physiologic down-regulation of T cell responsiveness in the face of persistent antigenic stimulation. In this study, we examined the role of CTLA-4 in this process using CTLA-4-deficient and wild-type TCR transgenic, Rag2(-/-), CD4(+) T cells transferred into a T cell-deficient, Ag-expressing host. Surprisingly, we found that the tuning process of adoptively transferred T cells could be induced and the hyporesponsive state maintained in the absence of CTLA-4. Furthermore, movement to a deeper state of anergy following restimulation in vivo in a second Ag-bearing host was also unaffected. In contrast, CTLA-4 profoundly inhibited late T cell expansion in vivo following both primary and secondary transfers, and curtailed IL-2 and IFN-gamma production. Removal of this braking function in CTLA-4-deficient mice following Ag stimulation may explain their lymphoproliferative dysregulation.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Antigens, CD
- Antigens, Differentiation/genetics
- Antigens, Differentiation/immunology
- Antigens, Differentiation/metabolism
- Antigens, Differentiation/physiology
- Antigens, Differentiation, T-Lymphocyte/biosynthesis
- CD3 Complex/genetics
- CD4-Positive T-Lymphocytes/cytology
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- CD4-Positive T-Lymphocytes/transplantation
- CTLA-4 Antigen
- Cell Division/genetics
- Cell Division/immunology
- Cell Proliferation
- Columbidae
- Cytochromes c/immunology
- Cytokines/antagonists & inhibitors
- Cytokines/biosynthesis
- Down-Regulation/genetics
- Down-Regulation/immunology
- Growth Inhibitors/deficiency
- Growth Inhibitors/genetics
- Growth Inhibitors/immunology
- Growth Inhibitors/metabolism
- Immunity, Innate/genetics
- Lymphocyte Activation/genetics
- Mice
- Mice, Inbred A
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Resting Phase, Cell Cycle/genetics
- Resting Phase, Cell Cycle/immunology
- Self Tolerance/genetics
Collapse
Affiliation(s)
- Manabu Inobe
- Laboratory of Cellular and Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | |
Collapse
|
598
|
Thompson RH, Gillett MD, Cheville JC, Lohse CM, Dong H, Webster WS, Krejci KG, Lobo JR, Sengupta S, Chen L, Zincke H, Blute ML, Strome SE, Leibovich BC, Kwon ED. Costimulatory B7-H1 in renal cell carcinoma patients: Indicator of tumor aggressiveness and potential therapeutic target. Proc Natl Acad Sci U S A 2004; 101:17174-9. [PMID: 15569934 PMCID: PMC534606 DOI: 10.1073/pnas.0406351101] [Citation(s) in RCA: 624] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2004] [Indexed: 12/15/2022] Open
Abstract
Expression of B7-H1, a costimulating glycoprotein in the B7 family, is normally restricted to macrophage-lineage cells, providing a potential costimulatory signal source for regulation of T cell activation. In contrast, aberrant expression of B7-H1 by tumor cells has been implicated in impairment of T cell function and survival, resulting in defective host antitumoral immunity. The relationship between tumor-associated B7-H1 and clinical cancer progression is unknown. Herein, we report B7-H1 expression by both renal cell carcinoma (RCC) tumors of the kidney and RCC tumor-infiltrating lymphocytes. In addition, our analysis of 196 clinical specimens reveals that patients harboring high intratumoral expression levels of B7-H1, contributed by tumor cells alone, lymphocytes alone, or tumor and/or lymphocytes combined, exhibit aggressive tumors and are at markedly increased risk of death from RCC. In fact, patients with high tumor and/or lymphocyte B7-H1 levels are 4.5 times more likely to die from their cancer than patients exhibiting low levels of B7-H1 expression (risk ratio 4.53; 95% confidence interval 1.94-10.56; P < 0.001.) Thus, our study suggests a previously undescribed mechanism whereby RCC may impair host immunity to foster tumor progression. B7-H1 may prove useful as a prognostic variable for RCC patients both pre- and posttreatment. In addition, B7-H1 may represent a promising target to facilitate more favorable responses in patients who require immunotherapy for treatment of advanced RCC.
Collapse
Affiliation(s)
- R Houston Thompson
- Department of Urology, Mayo Medical School, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
599
|
Abstract
Bone marrow-derived dendritic cells (DCs) are the most potent antigen-presenting cells capable of activating naïve T cells. Loading DCs ex vivo with tumor antigens can stimulate potent antitumor immunity in tumor-bearing mice. This review describes the use of mRNA-encoded tumor antigens as a form of antigen loaded onto DCs, including our early experience from clinical trials in urological cancers. Transfection of DCs with mRNA is simple and effective. Comparative studies suggest that mRNA transfection is superior to other antigen-loading techniques in generating immunopotent DCs. The ability to amplify RNA from microscopic amounts of tumor tissue extends the use of DC vaccination to virtually every cancer patient. The striking observation from two phase I clinical trials, in patients with prostate cancer immunized with prostate-specific antigen mRNA-transfected DCs and patients with renal cancer immunized with autologous tumor RNA-transfected DCs, was that the majority of patients exhibited a vaccine-induced T-cell response. Suggestive evidence of clinically related responses was seen in both the trials. Immunization with mRNA-transfected DCs is a promising strategy to stimulate potent antitumor immunity and could serve as a foundation for developing effective treatments for cancer.
Collapse
Affiliation(s)
- Eli Gilboa
- Department of Surgery, Center for Genetic and Cellular Therapies, Duke University Medical Center, Durham, NC 27710, USA.
| | | |
Collapse
|
600
|
Shahied LS, Tang Y, Alpaugh RK, Somer R, Greenspon D, Weiner LM. Bispecific Minibodies Targeting HER2/neu and CD16 Exhibit Improved Tumor Lysis When Placed in a Divalent Tumor Antigen Binding Format. J Biol Chem 2004; 279:53907-14. [PMID: 15471859 DOI: 10.1074/jbc.m407888200] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Unconjugated monoclonal antibodies have emerged as important therapeutic agents for selected malignancies. One mechanism by which antibodies can exert cytotoxic effects is antibody-dependent cellular cytotoxicity (ADCC). In an effort to increase the efficiency of ADCC at tumor sites, we have focused on the construction of bispecific antibodies specific for the tumor antigen HER2/neu and the Fc gamma RIII-activating receptor (CD16) found on NK cells, mononuclear phagocytes, and neutrophils. Here, we describe the production of bispecific minibodies in two distinct binding formats. The parent minibody was constructed such that the IgG1 C(H)3 constant domain serves as the oligomerization domain and is attached to an anti-CD16 and an anti-HER2/ neu single-chain Fv via 19- and 29-amino acid linkers, respectively. This molecule can be expressed in mammalian cells from a dicistronic vector and has been purified using sequential affinity purification techniques. Analysis by surface plasmon resonance shows that the bispecific minibody can bind to HER2/neu and CD16, both individually and simultaneously. Furthermore, cytotoxicity studies show that the minibody can induce significant tumor cell lysis at a concentration as low as 20 nm. A trimeric, bispecific minibody (TriBi) that binds dimerically to HER2/neu and monomerically to CD16 induces equivalent cytotoxicity at lower antibody concentrations than either the parent minibody or the corresponding single-chain dimer. Both minibody constructs are stable in mouse and human serum for up to 72 h at 37 degrees C. These minibodies have the potential to target solid tumors and promote tumor lysis by natural killer cells and mononuclear phagocytes.
Collapse
MESH Headings
- Animals
- Antibodies, Bispecific/administration & dosage
- Antibodies, Bispecific/genetics
- Antibodies, Bispecific/immunology
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/genetics
- Antibodies, Monoclonal/immunology
- Antibody Specificity
- Antibody-Dependent Cell Cytotoxicity
- Antigens, Neoplasm/immunology
- Binding Sites, Antibody
- Blood
- COS Cells
- Chlorocebus aethiops
- Cloning, Molecular
- Drug Stability
- Embryo, Mammalian
- Female
- Gene Expression
- Humans
- Immunoglobulin G/chemistry
- Immunoglobulin G/immunology
- Kidney
- Mice
- Neoplasms/immunology
- Ovarian Neoplasms
- Receptor, ErbB-2/immunology
- Receptors, IgG/immunology
- Transfection
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Lillian S Shahied
- Department of Medical Oncology, Fox Chase Cancer Center, Philadelphia, Pennsylvania 19111, USA
| | | | | | | | | | | |
Collapse
|