251
|
Enk AH, Becker JC, Schuler G. Immunotherapy of malignant melanoma - Basic principles and novel therapeutic approaches. J Dtsch Dermatol Ges 2006. [DOI: 10.1111/j.1610-0387.2005.05038.x-i1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
252
|
Speiser DE, Baumgaertner P, Barbey C, Rubio-Godoy V, Moulin A, Corthesy P, Devevre E, Dietrich PY, Rimoldi D, Liénard D, Cerottini JC, Romero P, Rufer N. A Novel Approach to Characterize Clonality and Differentiation of Human Melanoma-Specific T Cell Responses: Spontaneous Priming and Efficient Boosting by Vaccination. THE JOURNAL OF IMMUNOLOGY 2006; 177:1338-48. [PMID: 16818795 DOI: 10.4049/jimmunol.177.2.1338] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Despite major progress in T lymphocyte analysis in melanoma patients, TCR repertoire selection and kinetics in response to tumor Ags remain largely unexplored. In this study, using a novel ex vivo molecular-based approach at the single-cell level, we identified a single, naturally primed T cell clone that dominated the human CD8(+) T cell response to the Melan-A/MART-1 Ag. The dominant clone expressed a high-avidity TCR to cognate tumor Ag, efficiently killed tumor cells, and prevailed in the differentiated effector-memory T lymphocyte compartment. TCR sequencing also revealed that this particular clone arose at least 1 year before vaccination, displayed long-term persistence, and efficient homing to metastases. Remarkably, during concomitant vaccination over 3.5 years, the frequency of the pre-existing clone progressively increased, reaching up to 2.5% of the circulating CD8 pool while its effector functions were enhanced. In parallel, the disease stabilized, but subsequently progressed with loss of Melan-A expression by melanoma cells. Collectively, combined ex vivo analysis of T cell differentiation and clonality revealed for the first time a strong expansion of a tumor Ag-specific human T cell clone, comparable to protective virus-specific T cells. The observed successful boosting by peptide vaccination support further development of immunotherapy by including strategies to overcome immune escape.
Collapse
MESH Headings
- Antigen Presentation/immunology
- Antigens, Neoplasm
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/pathology
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/immunology
- Cell Differentiation/immunology
- Clone Cells
- Cytotoxicity Tests, Immunologic
- Disease Progression
- Epitopes, T-Lymphocyte/blood
- Epitopes, T-Lymphocyte/immunology
- Humans
- Immunization, Secondary
- Immunodominant Epitopes/administration & dosage
- Immunodominant Epitopes/immunology
- Lymphatic Metastasis/immunology
- Lymphatic Metastasis/pathology
- Lymphocyte Count
- MART-1 Antigen
- Melanoma/immunology
- Melanoma/pathology
- Melanoma/secondary
- Melanoma/therapy
- Neoplasm Proteins/blood
- Neoplasm Proteins/immunology
- Receptors, Antigen, T-Cell/analysis
- Receptors, Antigen, T-Cell/blood
- Receptors, Antigen, T-Cell/metabolism
- Time Factors
- Vaccines, Subunit/administration & dosage
- Vaccines, Subunit/immunology
Collapse
Affiliation(s)
- Daniel E Speiser
- Division of Clinical Onco-Immunology, Ludwig Institute for Cancer Research, University Hospital of Lausanne, Lausanne, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
253
|
Petrulio CA, Kim-Schulze S, Kaufman HL. The tumour microenvironment and implications for cancer immunotherapy. Expert Opin Biol Ther 2006; 6:671-84. [PMID: 16805707 DOI: 10.1517/14712598.6.7.671] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Tumour cells exist in a complex milieu of cellular and non-cellular components comprising fibroblasts, endothelial cells, immune cells and metabolites of cellular respiration. An elaborate interplay between these components and tumour cells exists with implications for immunological recognition of tumour cells. Tumours have been shown to alter their antigen and cytokine profiles, desensitise and impair immune defences, signal fibroblasts to facilitate metastasis, and take advantage of acidic and hypoxic conditions that impede normal cells. This paper aims to review the roles of the stroma, extracellular matrix and chemistry of the microenvironment on tumour growth, with particular emphasis on interactions with the immune system, and to highlight some of the novel therapeutic strategies that target the tumour microenvironment.
Collapse
Affiliation(s)
- Christian A Petrulio
- Columbia University, The Tumour Immunology Laboratory, 177 Fort Washington Avenue, MHB-7SK, New York, NY 10032, USA
| | | | | |
Collapse
|
254
|
Abstract
Many antigens recognized by autologous T lymphocytes have been identified on human melanoma. Melanoma patients usually mount a spontaneous T cell response against their tumor. But at some point, the responder T cells become ineffective, probably because of a local immunosuppressive process occurring at the tumor sites. Therapeutic vaccination of metastatic melanoma patients with these antigens is followed by tumor regressions only in a small minority of the patients. The T cell responses to the vaccines show correlation with the tumor regressions. The local immunosuppression may be the cause of the lack of vaccination effectiveness that is observed in most patients. In patients who do respond to the vaccine, the antivaccine T cells probably succeed in reversing focally this immunosuppression and trigger a broad activation of other antitumor T cells, which proceed to destroy the tumor.
Collapse
Affiliation(s)
- Thierry Boon
- Ludwig Institute for Cancer Research, Brussels Branch, and Cellular Genetics Unit, Université de Louvain, Brussels, Belgium.
| | | | | | | |
Collapse
|
255
|
Lizée G, Radvanyi LG, Overwijk WW, Hwu P. Immunosuppression in melanoma immunotherapy: potential opportunities for intervention. Clin Cancer Res 2006; 12:2359s-2365s. [PMID: 16609059 DOI: 10.1158/1078-0432.ccr-05-2537] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Although melanomas are substantially more immunogenic than other tumors, current immunotherapeutic approaches for melanoma patients have met with only limited success. Although melanoma-specific CD8+ T-cell responses can often be generated in patients naturally or through vaccination regimens, tumors frequently continue to grow unabated, suggesting that tumor-specific immune responses may be actively dampened in vivo. Research over the past decade has brought to light several mechanisms used by melanomas and other tumors to suppress tumor-specific immune responses. These include the presence of regulatory immune cells within the tumor microenvironment and draining lymph nodes that serve to shut down effector T-cell function. In addition, melanoma tumors themselves express a number of soluble and membrane-bound molecules that are responsible for inhibiting activated immune cells. The identification of these suppressive mechanisms has provided significant opportunities for designing novel therapeutic interventions that could augment current vaccination and adoptive transfer approaches for treatment of melanoma.
Collapse
Affiliation(s)
- Gregory Lizée
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | | | | | | |
Collapse
|
256
|
Overwijk WW, de Visser KE, Tirion FH, de Jong LA, Pols TWH, van der Velden YU, van den Boorn JG, Keller AM, Buurman WA, Theoret MR, Blom B, Restifo NP, Kruisbeek AM, Kastelein RA, Haanen JBAG. Immunological and antitumor effects of IL-23 as a cancer vaccine adjuvant. THE JOURNAL OF IMMUNOLOGY 2006; 176:5213-22. [PMID: 16621986 PMCID: PMC2242845 DOI: 10.4049/jimmunol.176.9.5213] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The promising, but modest, clinical results of many human cancer vaccines indicate a need for vaccine adjuvants that can increase both the quantity and the quality of vaccine-induced, tumor-specific T cells. In this study we tested the immunological and antitumor effects of the proinflammatory cytokine, IL-23, in gp100 peptide vaccine therapy of established murine melanoma. Neither systemic nor local IL-23 alone had any impact on tumor growth or tumor-specific T cell numbers. Upon specific vaccination, however, systemic IL-23 greatly increased the relative and absolute numbers of vaccine-induced CD8(+) T cells and enhanced their effector function at the tumor site. Although IL-23 specifically increased IFN-gamma production by tumor-specific T cells, IFN-gamma itself was not a primary mediator of the vaccine adjuvant effect. The IL-23-induced antitumor effect and accompanying reversible weight loss were both partially mediated by TNF-alpha. In contrast, local expression of IL-23 at the tumor site maintained antitumor activity in the absence of weight loss. Under these conditions, it was also clear that enhanced effector function of vaccine-induced CD8(+) T cells, rather than increased T cell number, is a primary mechanism underlying the antitumor effect of IL-23. Collectively, these results suggest that IL-23 is a potent vaccine adjuvant for the induction of therapeutic, tumor-specific CD8(+) T cell responses.
Collapse
Affiliation(s)
- Willem W Overwijk
- Division of Immunology, The Netherlands Cancer Institute, 1105 AZ Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
257
|
Vuylsteke RJCLM, Molenkamp BG, van Leeuwen PAM, Meijer S, Wijnands PGJTB, Haanen JBAG, Scheper RJ, de Gruijl TD. Tumor-Specific CD8+ T Cell Reactivity in the Sentinel Lymph Node of GM-CSF–Treated Stage I Melanoma Patients is Associated with High Myeloid Dendritic Cell Content. Clin Cancer Res 2006; 12:2826-33. [PMID: 16675577 DOI: 10.1158/1078-0432.ccr-05-2431] [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] [Indexed: 11/16/2022]
Abstract
PURPOSE Impaired immune functions in the sentinel lymph node (SLN) may facilitate early metastatic events during melanoma development. Local potentiation of tumor-specific T cell reactivity may be a valuable adjuvant treatment option. EXPERIMENTAL DESIGN We examined the effect of locally administered granulocyte/macrophage-colony stimulating factor (GM-CSF) on the frequency of tumor-specific CD8+ T cells in the SLN and blood of patients with stage I melanoma. Twelve patients were randomly assigned to preoperative local administration of either recombinant human GM-CSF or NaCl 0.9%. CD8+ T cells from SLN and peripheral blood were tested for reactivity in an IFNgamma ELISPOT assay against the full-length MART-1 antigen and a number of HLA-A1, HLA-A2, and HLA-A3-restricted epitopes derived from a range of melanoma-associated antigens. RESULTS Melanoma-specific CD8+ T cell response rates in the SLN were one of six for the control group and four of six for the GM-CSF-administered group. Only one patient had detectable tumor-specific CD8+ T cells in the blood, but at lower frequencies than in the SLN. All patients with detectable tumor-specific CD8+ T cells had a percentage of CD1a+ SLN-dendritic cells (DC) above the median (i.e., 0.33%). This association between above median CD1a+ SLN-DC frequencies and tumor antigen-specific CD8+ T cell reactivity was significant in a two-sided Fisher's exact test (P = 0.015). CONCLUSIONS Locally primed antitumor T cell responses in the SLN are detectable as early as stage I of melanoma development and may be enhanced by GM-CSF-induced increases in SLN-DC frequencies.
Collapse
Affiliation(s)
- Ronald J C L M Vuylsteke
- Department of Surgical Oncology, Pathology, VU University Medical Center, Amsterdam, the Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
258
|
Gattinoni L, Powell DJ, Rosenberg SA, Restifo NP. Adoptive immunotherapy for cancer: building on success. Nat Rev Immunol 2006; 6:383-93. [PMID: 16622476 PMCID: PMC1473162 DOI: 10.1038/nri1842] [Citation(s) in RCA: 655] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Adoptive cell transfer after host preconditioning by lymphodepletion represents an important advance in cancer immunotherapy. Here, we describe how a lymphopaenic environment enables tumour-reactive T cells to destroy large burdens of metastatic tumour and how the state of differentiation of the adoptively transferred T cells can affect the outcome of treatment. We also discuss how the translation of these new findings might further improve the efficacy of adoptive cell transfer through the use of vaccines, haematopoietic-stem-cell transplantation, modified preconditioning regimens, and alternative methods for the generation and selection of the T cells to be transferred.
Collapse
Affiliation(s)
- Luca Gattinoni
- National Cancer Institute, National Institutes of Health, Mark O. Hatfield Clinical Research Center, Room 3-5762, 10 Center Drive, Bethesda, Maryland 20892-1201, USA.
| | | | | | | |
Collapse
|
259
|
Velicu S, Han Y, Ulasov I, Brown IE, El Andaloussi A, Gajewski TF, Lesniak MS. Cross-priming of T cells to intracranial tumor antigens elicits an immune response that fails in the effector phase but can be augmented with local immunotherapy. J Neuroimmunol 2006; 174:74-81. [PMID: 16504307 DOI: 10.1016/j.jneuroim.2006.01.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2005] [Revised: 01/21/2006] [Accepted: 01/25/2006] [Indexed: 10/25/2022]
Abstract
Central nervous system (CNS) tumors are thought to be poorly immunogenic. However, whether defective anti-tumor immunity is a consequence of a relative failure of T cell priming versus a deficient effector phase of the anti-tumor immune response is not clear. We utilized a well-defined model system of B16 melanoma expressing the model antigen SIY-GFP to evaluate tumor antigen cross-priming and tumor rejection from the CNS versus subcutaneous compartments. We observed that B16-SIY cells implanted in the CNS were capable of inducing T cell priming as measured by IFN-gamma ELISPOT in the spleen. Cross-priming occurred in the absence of detectable systemic dissemination of the tumor. Despite the induction of a T cell response, CNS tumors grew progressively and were fatal, whereas the same tumor implanted in the flank was rejected. To study the effector phase of the immune response in more detail, in vitro primed 2C/RAG2-/- TCR transgenic CD8+ cells, which recognize the SIY peptide, were adoptively transferred. In addition, the CNS microenvironment was modulated by intracranial delivery of IL-2. While mice that received primed 2C cells alone showed an increase in survival, co-administration of intracranial IL-2 led to a marked prolongation of survival, with 20% of mice surviving at least 120 days. Our results demonstrate that CD8+ T cell cross-priming does indeed occur in response to a CNS tumor, but that manipulation of the brain tumor microenvironment may be necessary to support the effector phase of the anti-tumor immune response.
Collapse
Affiliation(s)
- Simona Velicu
- Division of Neurosurgery, The University of Chicago, Pritzker School of Medicine, 5841 S. Maryland Ave-MC 3026, Chicago, IL 60637, USA
| | | | | | | | | | | | | |
Collapse
|
260
|
Gajewski TF. Identifying and overcoming immune resistance mechanisms in the melanoma tumor microenvironment. Clin Cancer Res 2006; 12:2326s-2330s. [PMID: 16609053 DOI: 10.1158/1078-0432.ccr-05-2517] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The continually growing list of defined tumor antigens is broadening the potential applicability of tumor antigen-targeted cancer therapies. Although cancer vaccines and adoptive T-cell transfer have been shown to increase the frequency of circulating tumor antigen-specific T cells, these approaches cause clinical responses in a few patients. In melanoma, approximately one third of metastatic lesions contain activated T cells, including those specific for tumor antigens, arguing that the priming phase has occurred already in such individuals even without vaccination. These observations indicate that tumor resistance to immune destruction may dominate in many instances, arguing for a thorough analysis of the melanoma tumor microenvironment in individual patients. Recent work has suggested that T-cell anergy, the influence of CD4+ CD25+ regulatory T cells, the expression of inhibitory ligands, such as PD-L1, and the activity of nutrient-catabolizing enzymes, such as indoleamine 2,3-dioxygenase, may be involved. Preclinical murine models have shown that interfering with each of these processes can translate into T-cell-mediated tumor control. Importantly, each of these targets is amenable to clinical manipulation. Clinical translation of these approaches to counter negative regulation of antitumor immunity should receive high priority.
Collapse
Affiliation(s)
- Thomas F Gajewski
- Department of Pathology, University of Chicago, Chicago, Illinois 60637, USA.
| |
Collapse
|
261
|
Haanen JBAG, Baars A, Gomez R, Weder P, Smits M, de Gruijl TD, von Blomberg BME, Bloemena E, Scheper RJ, van Ham SM, Pinedo HM, van den Eertwegh AJM. Melanoma-specific tumor-infiltrating lymphocytes but not circulating melanoma-specific T cells may predict survival in resected advanced-stage melanoma patients. Cancer Immunol Immunother 2006; 55:451-8. [PMID: 16034562 PMCID: PMC11030194 DOI: 10.1007/s00262-005-0018-5] [Citation(s) in RCA: 108] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2005] [Accepted: 04/19/2005] [Indexed: 01/18/2023]
Abstract
PURPOSE To study the effect of autologous tumor cell vaccinations on the presence and numbers of circulating CD8+ T cells specific for tumor-associated antigens (TAA) in metastatic melanoma patients. To investigate the correlation between the presence of tumor-infiltrating lymphocytes (TIL) and circulating TAA-specific CD8+ T cells before and after autologous tumor cell vaccination with overall survival. EXPERIMENTAL DESIGN Twenty-five stage III and resected stage IV metastatic melanoma patients were adjuvantly treated with a series of intracutaneously injected autologous tumor cell vaccinations, of which the first two contained BCG as an immunostimulatory adjuvant. Tumor samples and blood samples obtained before and after vaccination of these patients were studied for the presence of TAA-specific T cells using HLA-tetramers and results were correlated with survival. RESULTS In 5 of 17 (29%) melanoma patients, circulating TAA-specific T cells were detectable prior to immunizations. No significant changes in the frequency and specificity were found during the treatment period in all patients. Presence of circulating TAA-specific T cells was not correlated with survival (log rank, P=0.215). Inside melanoma tissue, TAA-specific TIL could be detected in 75% of 16 available tumor samples. In case of detectable TAA-specific TIL, median survival was 22.5 months compared to median survival of 4.5 months in case of absence of TAA-specific T cells (log rank, P=0.0094). In none of the patients, TAA-specific T cells were found both in tumor tissue and blood at the same time. CONCLUSIONS These data suggest that the presence of TAA-specific TILs forms a prognostic factor, predicting improved survival in advanced-stage melanoma patients. The absence of TAA-specific T cells in the circulation suggests that homing of the tumor-specific T cell population to the tumor site contributes to the effectiveness of antitumor immunity.
Collapse
Affiliation(s)
- J. B. A. G. Haanen
- Division of Immunology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 Amsterdam, The Netherlands
| | - A. Baars
- Department of Medical Oncology, Vrije Universiteit Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands
| | - R. Gomez
- Division of Immunology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 Amsterdam, The Netherlands
| | - P. Weder
- Division of Immunology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 Amsterdam, The Netherlands
| | - M. Smits
- Division of Immunology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 Amsterdam, The Netherlands
| | - T. D. de Gruijl
- Department of Medical Oncology, Vrije Universiteit Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands
| | - B. M. E. von Blomberg
- Department of Pathology, Vrije Universiteit Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands
| | - E. Bloemena
- Department of Pathology, Vrije Universiteit Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands
| | - R. J. Scheper
- Department of Pathology, Vrije Universiteit Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands
| | - S. M. van Ham
- Department of Immunopathology, Sanquin Research at CLB, Plesmanlaan 125, 1066 Amsterdam, The Netherlands
| | - H. M. Pinedo
- , Vrije Universiteit Cancer Center, P.O.Box 7057, 1007 Amsterdam, The Netherlands
| | - A. J. M. van den Eertwegh
- Department of Medical Oncology, Vrije Universiteit Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands
| |
Collapse
|
262
|
Kudo-Saito C, Hodge JW, Kwak H, Kim-Schulze S, Schlom J, Kaufman HL. 4-1BB ligand enhances tumor-specific immunity of poxvirus vaccines. Vaccine 2006; 24:4975-86. [PMID: 16621183 PMCID: PMC1865095 DOI: 10.1016/j.vaccine.2006.03.042] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2006] [Revised: 03/14/2006] [Accepted: 03/16/2006] [Indexed: 10/24/2022]
Abstract
PURPOSE Recombinant poxvirus vaccines have been explored as tumor vaccines. The immunogenicity of these vaccines can be enhanced by co-expressing costimulatory molecules and tumor-associated antigens. While the B7-CD28 interaction has been most comprehensively investigated, other costimulatory molecules utilize different signaling pathways and might provide further cooperation in T cell priming and survival. 4-1BB (CD137) is a TNF family member and is critical for activation and long-term maintenance of primed T cells. This study was conducted to determine if a poxvirus expressing the ligand for 4-1BB (4-1BBL) could further improve the immune and therapeutic responses of a previously reported poxvirus vaccine expressing a triad of costimulatory molecules (B7.1, ICAM-1, and LFA-3). EXPERIMENTAL DESIGN A recombinant vaccinia virus expressing 4-1BBL was generated and characterized in an in vitro infection system. This vaccine was then used alone or in combination with a vaccinia virus expressing CEA, B7.1, ICAM-1, and LFA-3 in CEA-transgenic mice bearing established MC38 tumors. Tumor growth and immune responses against CEA and other tumor-associated antigens were determined. The level of anti-apoptotic proteins in responding T cells was determined by flow cytometry on tetramer selected T cells. RESULTS The combination of 4-1BBL with B7.1-based poxvirus vaccination resulted in significantly enhanced therapeutic effects against CEA-expressing tumors in a CEA-transgenic mouse model. This was associated with an increased level of CEA-specific CD4(+) and CD8(+) T cell responses, induction of antigen spreading to p53 and gp70, increased accumulation of CEA-specific T cells in the tumor microenvironment, and increased expression of bcl-X(L) and bcl-2 in CD4(+) and CD8(+) T cells in vaccinated mice. CONCLUSION 4-1BBL cooperates with B7 in enhancing anti-tumor and immunologic responses in a recombinant poxvirus vaccine model. The inclusion of costimulatory molecules targeting distinct T cell signaling pathways provides a mechanism for enhancing the therapeutic effectiveness of tumor vaccines.
Collapse
Affiliation(s)
- Chie Kudo-Saito
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892
| | - James W. Hodge
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892
| | - Heesun Kwak
- The Tumor Immunology Laboratory, Columbia University, 177 Fort Washington Avenue, New York, NY 10032
| | - Seunghee Kim-Schulze
- The Tumor Immunology Laboratory, Columbia University, 177 Fort Washington Avenue, New York, NY 10032
| | - Jeffrey Schlom
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892
| | - Howard L. Kaufman
- The Tumor Immunology Laboratory, Columbia University, 177 Fort Washington Avenue, New York, NY 10032
- To whom correspondence should be addressed, at The Tumor Immunology Laboratory, Columbia University, 177 Fort Washington Avenue, New York, NY 10032; E-mail:
| |
Collapse
|
263
|
Pittet MJ, Gati A, Le Gal FA, Bioley G, Guillaume P, de Smedt M, Plum J, Speiser DE, Cerottini JC, Dietrich PY, Romero P, Zippelius A. Ex vivo characterization of allo-MHC-restricted T cells specific for a single MHC-peptide complex. THE JOURNAL OF IMMUNOLOGY 2006; 176:2330-6. [PMID: 16455990 DOI: 10.4049/jimmunol.176.4.2330] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Alloreactive T cells are thought to be a potentially rich source of high-avidity T cells with therapeutic potential since tolerance to self-Ags is restricted to self-MHC recognition. Given the particularly high frequency of alloreactive T cells in the peripheral immune system, we used numerous MHC class I multimers to directly visualize and isolate viral and tumor Ag-specific alloreactive CD8 T cells. In fact, all but one specificities screened were undetectable in ex vivo labeling. In this study, we report the occurrence of CD8 T cells specifically labeled with allo-HLA-A*0201/Melan-A/MART-1(26-35) multimers at frequencies that are in the range of 10(-4) CD8 T cells and are thus detectable ex vivo by flow cytometry. We report the thymic generation and shaping of tumor Ag-specific, alloreactive T cells as well as their fate once seeded in the periphery. We show that these cells resemble their counterparts in HLA-A*0201-positive individuals, based on their structural and functional attributes.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Amino Acid Sequence
- Child
- Child, Preschool
- Histocompatibility Antigens/immunology
- Humans
- Infant
- Infant, Newborn
- Isoantigens/immunology
- Middle Aged
- Molecular Sequence Data
- Peptides/chemistry
- Peptides/immunology
- Phenotype
- Receptors, Antigen, T-Cell, alpha-beta/chemistry
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- Substrate Specificity
- T-Lymphocytes/immunology
- Transplantation, Homologous
Collapse
Affiliation(s)
- Mikaël J Pittet
- Division of Clinical Onco-Immunology, Ludwig Institute for Cancer Research, University Hospital, Lausanne, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
264
|
Joncker NT, Marloie MA, Chernysheva A, Lonchay C, Cuff S, Klijanienko J, Sigal-Zafrani B, Vincent-Salomon A, Sastre X, Lantz O. Antigen-independent accumulation of activated effector/memory T lymphocytes into human and murine tumors. Int J Cancer 2006; 118:1205-14. [PMID: 16152614 DOI: 10.1002/ijc.21472] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Tumor infiltrating lymphocytes (TIL) display activation markers and their presence is often associated with a favorable outcome. The role of tumor antigens in T cell recruitment into tumors is unclear. In an attempt to address this issue, we purified lymphocytes from breast tumor or nontumor, mammary tissue from patients, and normal mammary tissue from healthy individuals. In all groups, including healthy individuals, the majority of cells displayed an effector/memory (CD45RA(lo)/CD27(+/-)) phenotype and quite surprisingly the early and transient activation marker CD69, thus, questioning the tumor antigen specificity of TIL. Because the human repertoire is diverse, the T cells found in the tumors could recognize both self/tumor and environmental antigens through cross-reactivity. To test this hypothesis, we used two anti-male HY monospecific TCR transgenic mouse models. We found an infiltration of HY negative tumors by the CD4(+) and CD8(+) monoclonal T cells after priming with HY positive cells in the periphery. Thus, the presence of activated effector/memory T lymphocytes in tumors can be independent of reactivity against tumor antigens. These results suggest that to find activated effector T cells in a tissue does not always mean that a specific immune response is taking place.
Collapse
Affiliation(s)
- Nathalie T Joncker
- Laboratoire d'Immunologie, Institut Curie, 26 rue d'Ulm, 75005 Paris, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
265
|
Batard P, Peterson DA, Devêvre E, Guillaume P, Cerottini JC, Rimoldi D, Speiser DE, Winther L, Romero P. Dextramers: New generation of fluorescent MHC class I/peptide multimers for visualization of antigen-specific CD8+ T cells. J Immunol Methods 2006; 310:136-48. [PMID: 16516226 DOI: 10.1016/j.jim.2006.01.006] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2004] [Revised: 12/27/2005] [Accepted: 01/06/2006] [Indexed: 11/22/2022]
Abstract
Direct identification as well as isolation of antigen-specific T cells became possible since the development of "tetramers" based on avidin-fluorochrome conjugates associated with mono-biotinylated class I MHC-peptide monomeric complexes. In principle, a series of distinct class I MHC-peptide tetramers, each labelled with a different fluorochrome, would allow to simultaneously enumerate as many unique antigen-specific CD8(+) T cells. Practically, however, only phycoerythrin and allophycocyanin conjugated tetramers have been generally available, imposing serious constraints for multiple labeling. To overcome this limitation, we have developed dextramers which are multimers based on a dextran backbone bearing multiple fluorescein and streptavidin moieties. Here we demonstrate the functionality and optimization of these new probes on human CD8(+) T cell clones with four independent antigen specificities. Their applications to the analysis of relatively low frequency antigen-specific T cells in peripheral blood, as well as their use in fluorescence microscopy, are demonstrated. The data show that dextramers produce a stronger signal than their fluoresceinated tetramer counterparts. Thus, these could become the reagents of choice as the antigen-specific T cell labeling transitions from basic research to clinical application.
Collapse
Affiliation(s)
- Pascal Batard
- Division of Clinical Onco-Immunology, Ludwig Institute for Cancer Research, Lausanne Branch, University Hospital (CHUV), Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
266
|
Letsch A, Keilholz U, Kern F, Asemissen AM, Thiel E, Scheibenbogen C. Specific Central Memory T Cells in the Bone Marrow of Patients Immunized Against Tyrosinase Peptides. J Immunother 2006; 29:201-7. [PMID: 16531820 DOI: 10.1097/01.cji.0000180903.73965.72] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The goal of vaccination against tumors is the induction of effector T cells mediating tumor destruction and memory T cells providing long-term immunity. Several previous studies in patients vaccinated with major histocompatibility complex (MHC) class I peptides failed to show induction of central memory T cells, which are considered important to provide long-term memory. This study examined the subset composition and function of specific T cells generated by immunization with MHC class I binding tyrosinase peptides in combination with the adjuvants granulocyte-macrophage colony-stimulating factor and keyhole limpet hemocyanin in peripheral blood (PB) and bone marrow (BM) of melanoma patients. Most of the tyrosinase-specific T cells in PB had a CD45RA(+)CCR7(-) effector phenotype. In contrast to this, a large subset of tyrosinase-specific T cells in BM were memory T cells, including CD45RA(+)CCR7(-) central and CD45RA(-)CCR7(-) effector memory T cells. BM tyrosinase-specific T cells were functional, because they produced interferon-gamma and had a high proliferative potential. This study suggests that peptide vaccination can generate a fully functional memory T-cell response characterized by central and effector memory phenotypes, proliferative potential, and BM tropism.
Collapse
Affiliation(s)
- Anne Letsch
- Department of Hematology, Oncology, and Transfusion Medicine, Charité, Campus Benjamin Franklin, Berlin, Germany
| | | | | | | | | | | |
Collapse
|
267
|
Hirano N, Butler MO, Xia Z, Ansén S, von Bergwelt-Baildon MS, Neuberg D, Freeman GJ, Nadler LM. Engagement of CD83 ligand induces prolonged expansion of CD8+ T cells and preferential enrichment for antigen specificity. Blood 2006; 107:1528-36. [PMID: 16239433 PMCID: PMC1895397 DOI: 10.1182/blood-2005-05-2073] [Citation(s) in RCA: 139] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2005] [Accepted: 10/11/2005] [Indexed: 12/21/2022] Open
Abstract
Following T-cell receptor and CD28 signaling, CD8+ T cells express a receptor for CD83, a molecule up-regulated on functionally mature dendritic cells. Although this expression pattern suggests that CD83 is involved in adaptive immunity, little is known about its function in the periphery, and the existence of its ligand on T cells is controversial. We demonstrate that the engagement of the CD83 ligand (CD83L) preferentially enriches and significantly amplifies the number of antigen-specific CD8+ T cells. Coengagement of the T-cell receptor, CD28, and CD83L supports priming of naive CD8+ T cells that retain antigen specificity and cytotoxic function for more than 6 months. Therefore, engagement of the CD83L provides a unique signal to activated CD8+ T cells that could be exploited to generate long-lived antigen-specific cytotoxic T cells for the treatment of cancer and infection.
Collapse
Affiliation(s)
- Naoto Hirano
- Department of Medical Oncology, Dana-Farber Cancer Institute, 44 Binney St, Boston, MA 02115, USA.
| | | | | | | | | | | | | | | |
Collapse
|
268
|
Teague RM, Sather BD, Sacks JA, Huang MZ, Dossett ML, Morimoto J, Tan X, Sutton SE, Cooke MP, Ohlén C, Greenberg PD. Interleukin-15 rescues tolerant CD8+ T cells for use in adoptive immunotherapy of established tumors. Nat Med 2006; 12:335-41. [PMID: 16474399 DOI: 10.1038/nm1359] [Citation(s) in RCA: 186] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2005] [Accepted: 01/03/2006] [Indexed: 01/16/2023]
Abstract
CD8+ T cells can mediate eradication of established tumors, and strategies to amplify tumor-reactive T-cell numbers by immunization or ex vivo expansion followed by adoptive transfer are currently being explored in individuals with cancer. Generating effective CD8+ T cell-mediated responses to tumors is often impeded by T-cell tolerance to relevant tumor antigens, as most of these antigens are also expressed in normal tissues. We examined whether such tolerant T cells could be rescued and functionally restored for use in therapy of established tumors. We used a transgenic T-cell receptor (TCR) mouse model in which peripheral CD8+ T cells specific for a candidate tumor antigen also expressed in liver are tolerant, failing to proliferate or secrete interleukin (IL)-2 in response to antigen. Molecular and cellular analysis showed that these tolerant T cells expressed the IL-15 receptor alpha chain, and could be induced to proliferate in vitro in response to exogenous IL-15. Such proliferation abrogated tolerance and the rescued cells became effective in treating leukemia. Therefore, high-affinity CD8+ T cells are not necessarily deleted by encounter with self-antigen in the periphery, and can potentially be rescued and expanded for use in tumor immunotherapy.
Collapse
Affiliation(s)
- Ryan M Teague
- Department of Immunology, University of Washington School of Medicine, Office H-564 HSC, Box 357650, Seattle, Washington 98195, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
269
|
Khazaie K, von Boehmer H. The impact of CD4+CD25+ Treg on tumor specific CD8+ T cell cytotoxicity and cancer. Semin Cancer Biol 2006; 16:124-36. [PMID: 16443370 DOI: 10.1016/j.semcancer.2005.11.006] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
There is sufficient evidence to suggest that tumor growth elicits specific immune responses, including CD8(+) and CD4(+) T cell responses that may delay tumor growth and could potentially be harnessed to eradicate cancer. Nevertheless the frequent outcome of cancer is lethality associated with uncontrolled growth and dissemination of tumor cells. The failure of the immune response may be naturally programmed and related to a specific subpopulation of CD4(+)CD25(+) regulatory T cells, whose function is to protect us against autoimmunity. Recent investigations have shed light on the in vivo behavior and functions of these cells. It is becoming evident that a major impact of these cells is on the cytolytic action of specific CD8(+) T cells that target the tumor. Inhibition of cytotoxicity is dependent on TGF-beta signaling by the effector cells. Thus, targeting immune regulation may provide a promising approach to the immune therapy of cancer. This approach however could also have unexpected deleterious consequences, as surprising new observations indicate that regulatory T cells can also delay tumor growth by independent mechanisms that relate to their cross talk with the innate immune response to cancer.
Collapse
Affiliation(s)
- Khashayarsha Khazaie
- Department of Cancer Immunology and AIDS, Dana Farber Cancer Institute, Harvard Medical School, 44 Binney Street, Boston, MA 02115, USA.
| | | |
Collapse
|
270
|
Egorov IK. Mouse models of efficient and inefficient anti-tumor immunity, with emphasis on minimal residual disease and tumor escape. Cancer Immunol Immunother 2006; 55:1-22. [PMID: 16091932 PMCID: PMC11030122 DOI: 10.1007/s00262-005-0007-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2005] [Accepted: 03/25/2005] [Indexed: 10/25/2022]
Abstract
Tumor escape from the host immune response remains the major problem holding the development of immunotherapies for cancer. In this review, congenic mouse lines are discussed that differ dramatically in their ability to respond to tumors tested and, thereby, to survive or to succumb to the tumor and/or its metastases. This ability is under the control of either MHC class I or nontrivial MHC class II beta genes expressed in a small subpopulation of antigen-presenting cells. Two hypotheses can explain the results obtained so far: (1) emergence of tumor cell variants that escape the host immune response in morbid mice but are eliminated in survivors, and (2) tumor-induced immunosuppression, which is either efficient or not, depending on the congenic line used. It is argued that further experimentation on these congenics will allow to choose the correct hypothesis, and to characterize the mechanism(s) of elimination of minimal residual disease and prevention of tumor escape by the immune system of survivors as well as the reason(s) for its failure in morbid mice. It is also argued that the use of these models will substantially increase the chance to resolve the controversy of poor correlation of immunotherapy testing in mice with clinical results.
Collapse
Affiliation(s)
- Igor K Egorov
- The Jackson Laboratory, Bar Harbor, ME 04609-1500, USA,
| |
Collapse
|
271
|
Johnson TM, Sondak VK, Bichakjian CK, Sabel MS. The role of sentinel lymph node biopsy for melanoma: evidence assessment. J Am Acad Dermatol 2005; 54:19-27. [PMID: 16384752 DOI: 10.1016/j.jaad.2005.09.029] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2005] [Revised: 08/23/2005] [Accepted: 09/13/2005] [Indexed: 02/06/2023]
Affiliation(s)
- Timothy M Johnson
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, Michigan, USA.
| | | | | | | |
Collapse
|
272
|
Walter S, Bioley G, Bühring HJ, Koch S, Wernet D, Zippelius A, Pawelec G, Romero P, Stevanović S, Rammensee HG, Gouttefangeas C. High frequencies of functionally impaired cytokeratin 18-specific CD8+ T cells in healthy HLA-A2+ donors. Eur J Immunol 2005; 35:2876-85. [PMID: 16134083 DOI: 10.1002/eji.200526207] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Combining cell surface phenotyping with functional analysis, human CD8+ T cells have been divided into several subsets which are being studied extensively in diverse physiological situations, such as viral infection, cancer and ageing. In particular, so-called terminally differentiated effector cells possess a CD45RA+ CCR7- CD27- CD28- phenotype, contain perforin and, in different models, have been shown to exert direct ex vivo killing and to release interleukins upon both antigen-nonspecific and -specific stimulation. Using HLA class I multimers, we have identified a high frequency of peripheral CD8+ T cells that recognize a peptide derived from the self protein cytokeratin 18 presented by the HLA-A*0201 molecule. These cells can be detected in approximately 15% of the HLA-A2-positive healthy donors tested. A detailed analysis revealed that they must have divided extensively in vivo, have an effector cell phenotype and express various natural killer cell-associated receptors. Interestingly, however, they remained unresponsive to antigen-specific stimulation in vitro in terms of cytotoxicity and cytokine secretion. Thus, cytokeratin 18-specific cells constitute a frequently encountered, new CD8+ T lymphocyte subpopulation without classical effector status and with so far unknown function.
Collapse
Affiliation(s)
- Steffen Walter
- Department of Immunology, Institute for Cell Biology, Eberhard Karls University, Tübingen, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
273
|
Li Y, Bleakley M, Yee C. IL-21 influences the frequency, phenotype, and affinity of the antigen-specific CD8 T cell response. THE JOURNAL OF IMMUNOLOGY 2005; 175:2261-9. [PMID: 16081794 DOI: 10.4049/jimmunol.175.4.2261] [Citation(s) in RCA: 224] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
IL-21, a newly described cytokine belonging to the IL-2 gamma-chain receptor cytokine family (that includes IL-2, IL-7, and IL-15), has been described as an important regulator of the cellular immune response. In this study, the role of IL-21 in the generation of a human Ag-specific CD8+ T cell response is characterized by tracking a rare, but measurable population of self-Ag-specific T cells in vitro. Autologous dendritic cells pulsed with the melanoma antigen recognized T cells 1 self-peptide were used to stimulate CD8+ T cells from HLA-A2+ healthy donors and melanoma patients. We demonstrate that exposure to IL-21 increased the total number of MART-1-specific CD8+ T cells that could be elicited by >20-fold and, at the clonal level, enriched for a population of high-affinity CD8+ T cells with a peptide dose requirement more than 1 log(10)-fold less than their untreated counterparts. Phenotypic analysis of T cells from IL-21-treated cultures revealed a unique population of CD45RO+ CD28(high) CD8+ T cells, a phenotype that was stable for at least 4 wk after IL-21 exposure. These CD28(high) CD8+ T cells produced IL-2 upon Ag stimulation and represent potential helper-independent CTLs. Our studies demonstrate a significant role for IL-21 in the primary Ag-specific human CTL response and support the use of IL-21 in the ex vivo generation of potent Ag-specific CTLs for adoptive therapy or as an adjuvant cytokine during in vivo immunization against tumor Ags.
Collapse
Affiliation(s)
- Yongqing Li
- Program in Immunology, Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | | | | |
Collapse
|
274
|
Kusmartsev S, Nagaraj S, Gabrilovich DI. Tumor-associated CD8+ T cell tolerance induced by bone marrow-derived immature myeloid cells. THE JOURNAL OF IMMUNOLOGY 2005; 175:4583-92. [PMID: 16177103 PMCID: PMC1350970 DOI: 10.4049/jimmunol.175.7.4583] [Citation(s) in RCA: 245] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
T cell tolerance is a critical element of tumor escape. However, the mechanism of tumor-associated T cell tolerance remains unresolved. Using an experimental system utilizing the adoptive transfer of transgenic T cells into naive recipients, we found that the population of Gr-1+ immature myeloid cells (ImC) from tumor-bearing mice was able to induce CD8+ T cell tolerance. These ImC accumulate in large numbers in spleens, lymph nodes, and tumor tissues of tumor-bearing mice and are comprised of precursors of myeloid cells. Neither ImC from control mice nor progeny of tumor-derived ImC, including tumor-derived CD11c+ dendritic cells, were able to render T cells nonresponsive. ImC are able to take up soluble protein in vivo, process it, and present antigenic epitopes on their surface and induce Ag-specific T cell anergy. Thus, this is a first demonstration that in tumor-bearing mice CD8+ T cell tolerance is induced primarily by ImC that may have direct implications for cancer immunotherapy.
Collapse
Affiliation(s)
| | | | - Dmitry I. Gabrilovich
- Address for correspondence: Dmitry Gabrilovich MD, PhD H. Lee Moffitt Cancer Center, University of South Florida, MRC-2, Room 2067, 12902 Magnolia Dr. Tampa, FL, 33612. Ph. 813 903 6863; FAX 813 632 1328,
| |
Collapse
|
275
|
Sakamoto T, Saito H, Tatebe S, Tsujitani S, Ozaki M, Ito H, Ikeguchi M. Interleukin-10 expression significantly correlates with minor CD8+T-cell infiltration and high microvessel density in patients with gastric cancer. Int J Cancer 2005; 118:1909-14. [PMID: 16287061 DOI: 10.1002/ijc.21598] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
We aimed to investigate the relationships between interleukin-10 (IL-10) expression and both the clinicopathological findings and prognoses in patients with gastric cancer and to compare IL-10 expression with microvessel (MV) density and CD8+ T lymphocyte infiltration to evaluate its effects on angiogenesis and immune responses in gastric cancer. IL-10 expression was determined in gastric cancer patients by reverse transcription-polymerase chain reaction (RT-PCR) or immunohistochemical procedures. Two of 7 normal gastric tissues showed IL-10 mRNA expression, while its expressions were confirmed in all advanced gastric carcinoma tissues examined (n=11) by RT-PCR. Immunohistochemical staining demonstrated that IL-10 expression was detected in 52 (47.7%) of 109 cases. There was a close correlation between IL-10 expression and MV density. IL-10 expression inversely correlated with CD8+ T-lymphocyte infiltration. The prognoses of patients whose tumors expressed IL-10 were significantly worse than those of patients whose tumors did not express IL-10. Multivariate analysis indicated IL-10 expression was an independent prognostic factor. IL-10 might be associated with tumor progression by stimulating angiogenesis and suppressing immune responses in gastric cancer.
Collapse
Affiliation(s)
- Teruhisa Sakamoto
- Department of Surgery, Division of Surgical Oncology, Tottori University School of Medicine, Yonago 683-8504, Japan
| | | | | | | | | | | | | |
Collapse
|
276
|
Badoual C, Vingert B, Agueznay N, Adotevi O, Haicheur N, Molina T, Bruneval P, Fridman WH, Tartour E. [Phenotypic and functional analysis of T lymphocytes in cancer patients]. Ann Pathol 2005; 25:211-9. [PMID: 16230947 DOI: 10.1016/s0242-6498(05)80112-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
In preclinical tumor model and in human cancer, tumor antigen specific T lymphocytes play a key role in the control of tumor development. Nevertheless in numerous cases, the infiltrating tumor T cells do not seem to influence the clinical progression of the tumor. A better phenotypic and functional characterization of T cells in close contact with tumor associated with a comprehensive analysis of tumor evasion mechanism to the host response should lead to an optimization of cancer immunotherapy protocols.
Collapse
Affiliation(s)
- Cécile Badoual
- INSERM U 255, Institut Biomédical des Cordeliers, Université Paris 5
| | | | | | | | | | | | | | | | | |
Collapse
|
277
|
Mocellin S, Marincola FM, Young HA. Interleukin-10 and the immune response against cancer: a counterpoint. J Leukoc Biol 2005; 78:1043-51. [PMID: 16204623 DOI: 10.1189/jlb.0705358] [Citation(s) in RCA: 281] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Although interleukin-10 (IL-10) is commonly regarded as an anti-inflammatory, immunosuppressive cytokine that favors tumor escape from immune surveillance, a wealth of evidence is accumulating that IL-10 also possesses some immunostimulating properties. In fact, IL-10 has the pleiotropic ability of influencing positively and negatively the function of innate and adaptive immunity in different experimental models, which makes it questionable to merely categorize this cytokine as a target of anti-immune escape therapeutic strategies or rather, as an immunological adjuvant in the fight against cancer. Here, we review available data about the immunostimulating anticancer properties of IL-10, and in particular, we focus on the hypothesis that in contrast to what occurs in secondary lymphoid organs, IL-10 overexpression within the tumor microenvironment may catalyze cancer immune rejection.
Collapse
Affiliation(s)
- Simone Mocellin
- Department of Oncological & Surgical Sciences, University of Padova, Italy.
| | | | | |
Collapse
|
278
|
Ozenci V, Miller AM, Palmborg A, Egevad L, Jaremko GA, Kälkner KM, Pisa P. Presence and specificity of tumor associated lymphocytes from ascites fluid in prostate cancer. Prostate 2005; 65:20-6. [PMID: 15800933 DOI: 10.1002/pros.20229] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
BACKGROUND In the present study, we had the rare opportunity to study immunological responses of TAL from ascites fluid in a patient with hormone-refractory prostate cancer. METHODS We evaluated tumor antigen-specific T-cell responses, induced by either prostate specific antigen (PSA) pulsed dendritic cells (DCs) or PSA peptides, in TAL and peripheral blood lymphocytes. RESULTS DC stimulation with PSA protein induced recognition of naturally processed PSA epitopes by both blood and ascites T cells. In contrast, only ascites T cells recognized the PSA-3 peptide, after stimulation with PSA-pulsed DCs or peptides. Finally, although IFNgamma secreting T cells were detectable in both blood and ascites by ELISPOT, multiplex cytokine assay detected the presence of predominantly Th2 cytokines. CONCLUSIONS Although tumor antigen-specific TAL were detected in ascites fluid, these cells were producing immunosuppressive cytokines which may contribute to tumor escape from recognition and/or destruction by the immune system.
Collapse
Affiliation(s)
- Volkan Ozenci
- Immune and Gene Therapy Laboratory, Cancer Centre Karolinska, Karolinska Institute, Stockholm, Sweden
| | | | | | | | | | | | | |
Collapse
|
279
|
Speiser DE, Romero P. Toward improved immunocompetence of adoptively transferred CD8+ T cells. J Clin Invest 2005; 115:1467-9. [PMID: 15931384 PMCID: PMC1137014 DOI: 10.1172/jci25427] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Adoptive transfer of autologous or allogenic T cells to patients is being used with increased frequency as a therapy for infectious diseases and cancer. However, many questions remain with regard to defining optimized procedures for preparation and selection of T cell populations for transfer. In a new study in this issue of the JCI, Gattinoni and colleagues used a TCR transgenic mouse model to examine in vitro-generated tumor antigen-specific CD8+ T cells at various stages of differentiation for their efficacy in adoptive immunotherapy against transplantable melanoma. The results confirm that CD8+ T cells progressively lose immunocompetence with prolonged in vitro cultivation and suggest that effector CD8+ T cells alone may be considerably less potent at protecting hosts with advanced tumors than are less differentiated T cells.
Collapse
Affiliation(s)
- Daniel E Speiser
- Ludwig Institute for Cancer Research, Division of Clinical Onco-Immunology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland.
| | | |
Collapse
|
280
|
|
281
|
Nestle FO, Farkas A, Conrad C. Dendritic-cell-based therapeutic vaccination against cancer. Curr Opin Immunol 2005; 17:163-9. [PMID: 15766676 DOI: 10.1016/j.coi.2005.02.003] [Citation(s) in RCA: 115] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Early clinical trials, in which over 1000 cancer patients received dendritic cell (DC) vaccines, tested different vaccine preparations, but they did not always induce sufficient acquired immunity or meet the expected level of tumor regressions. Current studies aim to improve the DC vaccine approach and capture the potential of these cells in order to gain access to lymphoid tissues and induce strong cell-mediated immunity. DC clinical trials are moving towards a more professional environment, in accordance with the latest quality standards. This explains the current need for innovative well designed trials with defined endpoints that induce robust anti-tumor immunity.
Collapse
Affiliation(s)
- Frank O Nestle
- Department of Dermatology, University Hospital of Zurich, Gloriastrasse 31, 8091 Zurich, Switzerland.
| | | | | |
Collapse
|
282
|
Miller AM, Ozenci V, Kiessling R, Pisa P. Immune Monitoring in a Phase 1 Trial of a PSA DNA Vaccine in Patients with Hormone-Refractory Prostate Cancer. J Immunother 2005; 28:389-95. [PMID: 16000958 DOI: 10.1097/01.cji.0000165353.19171.41] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Prostate cancer remains a leading cause of cancer illness and death among men in Europe. No curative treatment exists when the disease has spread beyond the prostate. Immunotherapy with DNA vaccines has emerged as a potential therapeutic approach for the induction of antitumor specific cytotoxic T lymphocytes. In this study six patients with hormone-refractory prostate cancer were monitored for their ability to mount PSA-specific cellular responses after receiving a pVAX/PSA DNA vaccine (patients 1-3, 100 microg; patients 7-9, 900 microg) with recombinant GM-CSF and IL-2 as adjuvants. IFNgamma ELISPOT showed that naturally processed PSA protein and PSA peptides are recognized by T cells in the blood of some prostate cancer patients after a PSA DNA vaccine. Analysis of other cytokines showed the production of IL-4 and IL-6 but importantly did not show an increase in the number of IL-10-producing cells after vaccination in any of the patients. The authors conclude that a pVAX/PSA DNA vaccine can induce PSA-specific cellular immune responses in patients with hormone-refractory prostate cancer, thus emphasizing the potential for PSA as a target molecule for the immunotherapy of prostate cancer.
Collapse
Affiliation(s)
- Ashley M Miller
- Immune and Gene Therapy Laboratory, Cancer Centre Karolinska, Karolinska Institute, 171 76 Stockholm, Sweden.
| | | | | | | |
Collapse
|
283
|
Lucignani G, Rescigno M. The role of molecular imaging in the development of dendritic cell-based cancer vaccines. Eur J Nucl Med Mol Imaging 2005; 32:725-30. [PMID: 15995874 DOI: 10.1007/s00259-005-1858-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
284
|
Kaufman HL, Deraffele G, Mitcham J, Moroziewicz D, Cohen SM, Hurst-Wicker KS, Cheung K, Lee DS, Divito J, Voulo M, Donovan J, Dolan K, Manson K, Panicali D, Wang E, Hörig H, Marincola FM. Targeting the local tumor microenvironment with vaccinia virus expressing B7.1 for the treatment of melanoma. J Clin Invest 2005; 115:1903-12. [PMID: 15937544 PMCID: PMC1142116 DOI: 10.1172/jci24624] [Citation(s) in RCA: 98] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2005] [Accepted: 03/31/2005] [Indexed: 12/15/2022] Open
Abstract
Immunotherapy for the treatment of metastatic melanoma remains a major clinical challenge. The melanoma microenvironment may lead to local T cell tolerance in part through downregulation of costimulatory molecules, such as B7.1 (CD80). We report the results from the first clinical trial, to our knowledge, using a recombinant vaccinia virus expressing B7.1 (rV-B7.1) for monthly intralesional vaccination of accessible melanoma lesions. A standard 2-dose-escalation phase I clinical trial was conducted with 12 patients. The approach was well tolerated with only low-grade fever, myalgias, and fatigue reported and 2 patients experiencing vitiligo. An objective partial response was observed in 1 patient and disease stabilization in 2 patients, 1 of whom is alive without disease 59 months following vaccination. All patients demonstrated an increase in postvaccination antibody and T cell responses against vaccinia virus. Systemic immunity was tested in HLA-A*0201 patients who demonstrated an increased frequency of gp100 and T cells specific to melanoma antigen recognized by T cells 1 (MART-1), also known as Melan-A, by ELISPOT assay following local rV-B7.1 vaccination. Local immunity was evaluated by quantitative real-time RT-PCR, which suggested that tumor regression was associated with increased expression of CD8 and IFN-gamma. The local delivery of vaccinia virus expressing B7.1 was well tolerated and represents an innovative strategy for altering the local tumor microenvironment in patients with melanoma.
Collapse
Affiliation(s)
- Howard L Kaufman
- Department of Surgery, Columbia University, New York, NY 10032, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
285
|
Coulie PG, Connerotte T. Human tumor-specific T lymphocytes: does function matter more than number? Curr Opin Immunol 2005; 17:320-5. [PMID: 15886124 DOI: 10.1016/j.coi.2005.03.002] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
In recent years, several clinical trials have involved the vaccination of cancer patients with tumor-specific antigens that are recognized by T lymphocytes. Anti-vaccine T-cell responses in these patients have been monitored on the assumption that their magnitude would correlate with clinical efficacy. Although analysis of these data show that such a correlation is emerging, detailed analyses of the few patients who benefit clinically from the vaccinations suggest that the function of the anti-vaccine T cells might be more important than their number. Recent studies show that in cancer patients numerous tumor-specific T cells appear to be quiescent in the presence of the tumor. Understanding how an efficient vaccine interferes with this coexistence is one of the current challenges of cancer immunotherapy.
Collapse
Affiliation(s)
- Pierre G Coulie
- Institute of Cellular Pathology, Université catholique de Louvain, Avenue Hippocrate 74, UCL 7459, B-1200 Brussels, Belgium.
| | | |
Collapse
|
286
|
Liu K, Caldwell SA, Abrams SI. Immune Selection and Emergence of Aggressive Tumor Variants as Negative Consequences of Fas-Mediated Cytotoxicity and Altered IFN-γ-Regulated Gene Expression. Cancer Res 2005; 65:4376-88. [PMID: 15899830 DOI: 10.1158/0008-5472.can-04-4269] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Antitumor responses can be induced in patients via active or adoptive immunotherapy, yet complete tumor eradication occurs infrequently. This paradox in tumor immunology led us to address two questions: (a) Does an antitumor response, which is intended to destroy the aberrant target population, also at the same time select for aggressive tumor variants (ATV) in vivo? (b) If this process does occur, what is the contribution of the perforin- or Fas-mediated effector mechanism in the immune selection of such ATV? Here, in an experimental mouse lung metastasis model, we showed that ATV generated either naturally in vivo or in vitro by anti-Fas selection resembled each other biologically and genetically as judged by enhanced tumor growth and genome-scale gene expression profiling, respectively. Furthermore, ATV that survived CTL adoptive immunotherapy displayed an even more profound loss of Fas expression and function as well as enhanced malignant proficiency in vivo. ATV, however, retained sensitivity to perforin-mediated lysis in vitro. Lastly, such ATV displayed a diminished responsiveness in their expression of IFN-gamma-regulated genes, including those mechanistically linked to Fas-mediated death (i.e., Fas and caspase-1). Overall, we showed that (a) immune selection did occur in vivo and played an important role in the emergence of ATV, (b) ATV bearing a Fas-resistant phenotype was a chief consequence of immune selection, and (c) an overall diminished responsiveness of IFN-gamma-regulated gene expression was characteristic of ATV. Thus, in this model, Fas-mediated cytotoxicity, in concert with IFN-gamma-regulated gene expression, mechanistically constituted significant determinants of immune selection of ATV in vivo.
Collapse
Affiliation(s)
- Kebin Liu
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland 20892-1402, USA
| | | | | |
Collapse
|
287
|
Slingluff CL, Speiser DE. Progress and controversies in developing cancer vaccines. J Transl Med 2005; 3:18. [PMID: 15862126 PMCID: PMC1142519 DOI: 10.1186/1479-5876-3-18] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2005] [Accepted: 04/29/2005] [Indexed: 11/10/2022] Open
Abstract
Immunotherapy has become a standard approach for cancer management, through the use of cytokines (eg: interleukin-2) and monoclonal antibodies. Cancer vaccines hold promise as another form of immunotherapy, and there has been substantial progress in identifying shared antigens recognized by T cells, in developing vaccine approaches that induce antigen-specific T cell responses in cancer patients, and in developing new technology for monitoring immune responses in various human tissue compartments. Dramatic clinical regressions of human solid tumors have occurred with some cancer vaccines, but the rate of those responses remains low. This article is part of a 2-part point:counterpoint series on peptide vaccines and adoptive therapy approaches for cancer. The current status of cancer vaccination, and associated challenges, are discussed. Emphasis is placed on the need to increase our knowledge of cancer immunobiology, as well as to improve monitoring of cellular immune function after vaccination. Progress in both areas will facilitate development of effective cancer vaccines, as well as of adoptive therapy. Effective cancer vaccines promise to be useful for treatment and prevention of cancer at low cost and with low morbidity.
Collapse
Affiliation(s)
| | - Daniel E Speiser
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
| |
Collapse
|
288
|
Chen DS, Davis MM. Cellular immunotherapy: antigen recognition is just the beginning. ACTA ACUST UNITED AC 2005; 27:119-27. [PMID: 15834723 DOI: 10.1007/s00281-005-0200-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2005] [Accepted: 02/14/2005] [Indexed: 11/26/2022]
Abstract
Advances in molecular and cellular biology have illustrated both the flexibility and complexity involved in host immune responses. Understanding this response is vital to the further development of therapeutic strategies that involve manipulation of the cellular immune response to target tumors. Mobilized, tumor antigen-specific T cells, the core for most immunotherapeutic strategies, are highly regulated, and capable of a wide spectrum of functional responses. Due to differences in murine and human immunity, broad-scale immune monitoring, particularly high-throughput ex vivo analysis of human immune responses, promises to determine what comprises an effective immunotherapy. Such understanding will lead to more sophisticated clinical trials, earlier determination of efficacy and individualized protocols.
Collapse
Affiliation(s)
- Daniel S Chen
- Department of Internal Medicine, Division of Oncology, Stanford University, Stanford, California 94305-5124, USA
| | | |
Collapse
|
289
|
Bronte V, Kasic T, Gri G, Gallana K, Borsellino G, Marigo I, Battistini L, Iafrate M, Prayer-Galetti T, Pagano F, Viola A. Boosting antitumor responses of T lymphocytes infiltrating human prostate cancers. ACTA ACUST UNITED AC 2005; 201:1257-68. [PMID: 15824085 PMCID: PMC2213151 DOI: 10.1084/jem.20042028] [Citation(s) in RCA: 289] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Immunotherapy may provide valid alternative therapy for patients with hormone-refractory metastatic prostate cancer. However, if the tumor environment exerts a suppressive action on antigen-specific tumor-infiltrating lymphocytes (TIL), immunotherapy will achieve little, if any, success. In this study, we analyzed the modulation of TIL responses by the tumor environment using collagen gel matrix–supported organ cultures of human prostate carcinomas. Our results indicate that human prostatic adenocarcinomas are infiltrated by terminally differentiated cytotoxic T lymphocytes that are, however, in an unresponsive status. We demonstrate the presence of high levels of nitrotyrosines in prostatic TIL, suggesting a local production of peroxynitrites. By inhibiting the activity of arginase and nitric oxide synthase, key enzymes of L-arginine metabolism that are highly expressed in malignant but not in normal prostates, reduced tyrosine nitration and restoration of TIL responsiveness to tumor were achieved. The metabolic control exerted by the tumor on TIL function was confirmed in a transgenic mouse prostate model, which exhibits similarities with human prostate cancer. These results identify a novel and dominant mechanism by which cancers induce immunosuppression in situ and suggest novel strategies for tumor immunotherapy.
Collapse
Affiliation(s)
- Vincenzo Bronte
- Department of Oncology and Surgical Sciences, University of Padova, 35100 Padova, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
290
|
Hambach L, Goulmy E. Immunotherapy of cancer through targeting of minor histocompatibility antigens. Curr Opin Immunol 2005; 17:202-10. [PMID: 15766682 DOI: 10.1016/j.coi.2005.01.010] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Minor histocompatibility antigens are allogeneic targets of T-cell mediated graft-versus-tumour effects following allogeneic stem cell transplantation. Recent research has identified several minor histocompatibility antigens as tumour proteins and has also disclosed their unique properties in both the induction and the effector phase of graft-versus-tumour effects. Targeting tumour-specific minor histocompatibility antigens by adoptive immunotherapy will battle against tumour tolerance and evoke allo-immune responses, thereby enhancing graft-versus-tumour effects against leukaemia and solid tumours. Recently acquired knowledge of the role of donor immunisation status, new techniques in the generation of minor histocompatibility antigen-specific cytotoxic T lymphocytes in vitro, and innovative principles in vaccination will help to design clinical trials that exploit minor histocompatibility antigens in the immunotherapy of cancer.
Collapse
Affiliation(s)
- Lothar Hambach
- Department of Immunohaematology and Bloodtransfusion, Leiden University Medical Center, Post Box 9600, 2300 RC Leiden, The Netherlands
| | | |
Collapse
|
291
|
Abstract
The discovery of defined tumor antigens and their application in therapeutic cancer vaccines has not yet resulted in a successful therapy for cancer patients. Recent data suggest that this might be because most current clinical immunotherapeutic strategies rely on a tolerized tumor-reactive T-cell repertoire, resulting in a weak T-cell response that cannot induce tumor regression in the face of a multitude of normal and tumor-induced immunoregulatory mechanisms. New insights from animal models and clinical trials suggest a rationale for combination approaches in which the ineffective endogenous anti-tumor immune response is enhanced through a combination of adoptive cell transfer (ACT), specific vaccination and cytokine help for the reliable induction of a robust anti-tumor immune response and tumor regression.
Collapse
Affiliation(s)
- Willem W Overwijk
- Department of Melanoma Medical Oncology, MD Anderson Cancer Center, 1515 Holcombe Boulevard, Box 904, Houston, Texas 77030, USA.
| |
Collapse
|
292
|
Germeau C, Ma W, Schiavetti F, Lurquin C, Henry E, Vigneron N, Brasseur F, Lethé B, De Plaen E, Velu T, Boon T, Coulie PG. High frequency of antitumor T cells in the blood of melanoma patients before and after vaccination with tumor antigens. ACTA ACUST UNITED AC 2005; 201:241-8. [PMID: 15657293 PMCID: PMC2212796 DOI: 10.1084/jem.20041379] [Citation(s) in RCA: 183] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
After vaccination of melanoma patients with MAGE antigens, we observed that even in the few patients showing tumor regression, the frequency of anti-vaccine T cells in the blood was often either undetectable or <10−5 of CD8 T cells. This frequency being arguably too low for these cells to be sole effectors of rejection, we reexamined the contribution of T cells recognizing other tumor antigens. The presence of such antitumor T cells in melanoma patients has been widely reported. To begin assessing their contribution to vaccine-induced rejection, we evaluated their blood frequency in five vaccinated patients. The antitumor cytotoxic T lymphocyte (CTL) precursors ranged from 10−4 to 3 × 10−3, which is 10–10,000 times higher than the anti-vaccine CTL in the same patient. High frequencies were also observed before vaccination. In a patient showing nearly complete regression after vaccination with a MAGE-3 antigen, we observed a remarkably focused antitumoral response. A majority of CTL precursors (CTLp's) recognized antigens encoded by MAGE-C2, another cancer-germline gene. Others recognized gp100 antigens. CTLp's recognizing MAGE-C2 and gp100 antigens were already present before vaccination, but new clonotypes appeared afterwards. These results suggest that a spontaneous antitumor T cell response, which has become ineffective, can be reawakened by vaccination and contribute to tumor rejection. This notion is reinforced by the frequencies of anti-vaccine and antitumor CTLs observed inside metastases, as presented by Lurquin et al. (Lurquin, C., B. Lethé, V. Corbière, I. Théate, N. van Baren, P.G. Coulie, and T. Boon. 2004. J. Exp. Med. 201:249–257).
Collapse
Affiliation(s)
- Catherine Germeau
- Cellular Genetics Unit, Université de Louvain, B-1200 Brussels, Belgium
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
293
|
Affiliation(s)
| | | | - Pedro Romero
- Ludwig Institute for Cancer ResearchLausanneSwitzerland
| |
Collapse
|
294
|
Chen ML, Pittet MJ, Gorelik L, Flavell RA, Weissleder R, von Boehmer H, Khazaie K. Regulatory T cells suppress tumor-specific CD8 T cell cytotoxicity through TGF-beta signals in vivo. Proc Natl Acad Sci U S A 2004; 102:419-24. [PMID: 15623559 PMCID: PMC544311 DOI: 10.1073/pnas.0408197102] [Citation(s) in RCA: 624] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Cancer patients can harbor significant numbers of CD8 and CD4 T cells with specificities to tumor antigens (Ags). Yet, in most cases, such T cells fail to eradicate the tumor in vivo. Here, we investigated the interference of Ag-specific CD4(+)CD25(+) regulatory T cells (Treg) with the tumor-specific CD8 T cell immune response in vivo, by monitoring the homing, expansion, and effector function of both subsets in draining and nondraining lymph nodes. The results show that CD8 cells expand to the same extent and produce similar levels of IFN-gamma in the presence or absence of Ag-specific Treg. Nevertheless, these Treg abrogate CD8 T cell-mediated tumor rejection by specifically suppressing the cytotoxicity of expanded CD8 cells. The molecular mechanism of suppression involves TGF-beta because expression of a dominant-negative TGF-beta receptor by tumor-specific CD8 cells renders them resistant to suppression and is associated with tumor rejection and unimpaired cytotoxicity.
Collapse
Affiliation(s)
- Mei-Ling Chen
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | |
Collapse
|
295
|
Milling SWF, Sai T, Silvers WK, Mintz B. Inhibition of melanoma growth after treatment with dendritic cells in a Tyr-SV40E murine model requires CD4+ T cells but not CD8+ T cells. Melanoma Res 2004; 14:555-62. [PMID: 15577330 DOI: 10.1097/00008390-200412000-00019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Melanomas are promising targets for immunotherapy, as they express a number of tissue-specific antigens against which immune responses can be elicited. We have previously described transgenic mice in which malignant cutaneous melanomas are produced. The 1042 melanoma cell line, derived from a primary melanoma in one of these mice, was used here to generate tumours by subcutaneous inoculation in syngeneic animals. All mice injected with 1 x 10(6) cells of the 1042 cell line developed a tumour. CD4+ T cells, CD8+ T cells and macrophages infiltrated the tumours. Treatment with dendritic cells pulsed with peptides from melanogenic proteins slowed tumour growth and resulted in increased numbers of infiltrating lymphocytes and macrophages, expansion of CD4+ T cells specific for 1042 cell antigens, and increased levels of 1042-specific immunoglobulin G1 (IgG1) and IgM in serum. The frequency of cytotoxic T lymphocytes (CTLs) specific for the MART-1 melanocytic antigen did not increase after dendritic cell treatment. Indeed, the presence of CD8+ T cells was apparently not required for the anti-tumour effects: slowing of tumour growth was not abrogated in animals depleted of CD8+ T cells using antibodies, or in syngeneic CD8-/- animals. In contrast, treatment with dendritic cells + peptides was ineffective after depletion of CD4+ T cells and in syngeneic CD4-/- mice. This experimental system therefore provides an opportunity to investigate CD4-dependent anti-tumour effector mechanisms, and for studies designed to activate the quiescent CTLs which infiltrate melanomas.
Collapse
MESH Headings
- Animals
- Antigens, Neoplasm
- CD4-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/immunology
- Cell Proliferation
- Cells, Cultured
- Cytokines/metabolism
- Cytotoxicity Tests, Immunologic
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Immunoglobulin G/blood
- Lymphocytes, Tumor-Infiltrating/immunology
- MART-1 Antigen
- Macrophages
- Melanoma, Experimental/immunology
- Melanoma, Experimental/pathology
- Melanoma, Experimental/therapy
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Monophenol Monooxygenase/genetics
- Monophenol Monooxygenase/immunology
- Monophenol Monooxygenase/metabolism
- Neoplasm Proteins/metabolism
- Peptide Fragments/immunology
- Peptide Fragments/metabolism
- Simian virus 40/genetics
- Simian virus 40/immunology
- Simian virus 40/metabolism
- Skin Neoplasms/immunology
- Skin Neoplasms/pathology
- Skin Neoplasms/therapy
- T-Lymphocytes, Cytotoxic
Collapse
Affiliation(s)
- Simon W F Milling
- Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia, PA, USA.
| | | | | | | |
Collapse
|
296
|
Roeder C, Schuler-Thurner B, Berchtold S, Vieth G, Driesch PVD, Schuler G, Lüftl M. MAGE-A3 is a frequent tumor antigen of metastasized melanoma. Arch Dermatol Res 2004; 296:314-9. [PMID: 15570431 DOI: 10.1007/s00403-004-0527-7] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2004] [Revised: 09/22/2004] [Accepted: 11/04/2004] [Indexed: 10/26/2022]
Abstract
MAGE-3 or MAGE-A3 is one of the best-characterized tumor antigens. Due to its tumor-restricted expression pattern and its recognition by both cytotoxic and helper T cells it constitutes a promising tumor antigen for anticancer immunotherapy, notably of malignant melanoma. Surprisingly, however, only very limited information is available on the frequency and consistency of its expression in metastatic melanoma lesions. We have now investigated the presence of MAGE-A3 mRNA in 316 tumor samples from 147 melanoma patients by RT-PCR. MAGE-A3 mRNA was detectable in 62% of metastases, and expression did not depend on the site of the metastases (skin, lymph node, and internal organs), age, sex, or duration of disease. Southern blot hybridization of the PCR product enhanced sensitivity of detection, and 26% more samples (13/50 samples tested) scored positive, indicating an even higher MAGE-A3 mRNA frequency than determined by simple ethidium bromide gel analysis. In 62 patients, we were able to investigate MAGE-A3 expression in several metastases from the same patient, and unexpectedly, both MAGE-A3-positive and MAGE-A3-negative metastases were found in 32% of these patients (20 of 62). Immunohistochemistry (using mAb 57B) demonstrated that the expression pattern was usually also heterogeneous with positively and negatively stained tumor cells within one metastasis. However, most (90%) of the metastases (47/52) gave a partially positive signal. Taken together, MAGE-A3 is a common and frequent tumor antigen in metastasized melanoma, but its expression is often heterogeneous.
Collapse
Affiliation(s)
- Claudia Roeder
- Department of Dermatology, University Hospital of Erlangen, Hartmannstr. 14, Erlangen, 91052, Germany
| | | | | | | | | | | | | |
Collapse
|
297
|
Meidenbauer N, Zippelius A, Pittet MJ, Laumer M, Vogl S, Heymann J, Rehli M, Seliger B, Schwarz S, Le Gal FA, Dietrich PY, Andreesen R, Romero P, Mackensen A. High Frequency of Functionally Active Melan-A–Specific T Cells in a Patient with Progressive Immunoproteasome-Deficient Melanoma. Cancer Res 2004; 64:6319-26. [PMID: 15342421 DOI: 10.1158/0008-5472.can-04-1341] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Abstract
Tumor-reactive T cells play an important role in cancer immunosurveillance. Applying the multimer technology, we report here an unexpected high frequency of Melan-A–specific CTLs in a melanoma patient with progressive lymph node metastases, consisting of 18 and 12.8% of total peripheral blood and tumor-infiltrating CD8+ T cells, respectively. Melan-A–specific CTLs revealed a high cytolytic activity against allogeneic Melan-A–expressing target cells but failed to kill the autologous tumor cells. Loading of the tumor cells with Melan-A peptide reversed the resistance to killing, suggesting impaired function of the MHC class I antigen processing and presentation pathway. Mutations of the coding region of the HLA-A2 binding Melan-A26–35 peptide or down-regulation of the MHC class I heavy chain, the antigenic peptide TAP, and tapasin could be excluded. However, PCR and immunohistochemical analysis revealed a deficiency of the immunoproteasomes low molecular weight protein 2 and low molecular weight protein 7 in the primary tumor cells, which affects the quantity and quality of generated T-cell epitopes and might explain the resistance to killing. This is supported by our data, demonstrating that the resistance to killing can be partially reversed by pre-exposure of the tumor cells to IFN-γ, which is known to induce the immunoproteasomes. Overall, this is the first report of an extremely high frequency of tumor-specific CTLs that exhibit competent T-cell–effector functions but fail to lyse the autologous tumor cells. Immunotherapeutic approaches should not only focus on the induction of a robust antitumor immune response, but should also have to target tumor immune escape mechanisms.
Collapse
Affiliation(s)
- Norbert Meidenbauer
- Department of Hematology/Oncology, University of Regensburg, Regensburg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
298
|
Mahnke YD, Speiser D, Luescher IF, Cerottini JC, Romero P. Recent advances in tumour antigen-specific therapy:In vivo veritas. Int J Cancer 2004; 113:173-8. [PMID: 15386403 DOI: 10.1002/ijc.20572] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Modern cancer therapies should strive not only to eliminate malignant tissues but also to preserve healthy tissues and the patient's quality of life. Antigen-specific immunotherapy approaches are promising for either aspect since they are designed to only act against tissues expressing 1 or more specified tumour antigens. In order to develop successful vaccine and adoptive transfer protocols, longitudinal monitoring of cancer patients taking part in clinical trials is mandatory. Here, in vivo expansion of antigen-specific cells, as well as their ex vivo functional status represent important parameters to be analysed. To obtain results that most closely reflect the cells' in vivo status, functional assays must be carried out with as little in vitro culturing as possible. The present minireview discusses recent advances in these domains.
Collapse
Affiliation(s)
- Yolanda D Mahnke
- Division of Clinical Onco-Immunology, Ludwig Institute for Cancer Research, University Hospital (CHUV), Avenue Pierre-Decker 4, 1005 Lausanne, Switzerland
| | | | | | | | | |
Collapse
|