51
|
Godet Y, Moreau-Aubry A, Mompelat D, Vignard V, Khammari A, Dreno B, Lang F, Jotereau F, Labarriere N. An additional ORF on meloe cDNA encodes a new melanoma antigen, MELOE-2, recognized by melanoma-specific T cells in the HLA-A2 context. Cancer Immunol Immunother 2010; 59:431-9. [PMID: 19730858 PMCID: PMC11029903 DOI: 10.1007/s00262-009-0762-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2009] [Accepted: 08/20/2009] [Indexed: 11/25/2022]
Abstract
We characterized a new melanoma antigen derived from one of the multiple open reading frames (ORFs) of the meloe transcript. The meloe gene is overexpressed in melanomas as compared to other cancer cell lines and normal tissues. The corresponding transcript is rather unusual, in that it does not contain a long unique ORF but multiple short ORFs. We recently characterized a tumor epitope derived from a polypeptide (MELOE-1) encoded by the ORF(1230-1370) and involved in relapse prevention of melanoma patients treated with autologous tumor infiltrating lymphocytes (TIL). Here we show that the ORF(285-404) encodes a polypeptide called MELOE-2 that also generated a HLA-A2 epitope recognized by a melanoma-specific T cell clone derived from the same TIL population from which we derived the MELOE-1-specific T cell clone. We also showed that HLA-A2 melanoma cells were spontaneously recognized by the MELOE-2-specific T cell clone, and we detected the presence of MELOE-2 reactive T cells in another TIL population infused to a patient who remained relapse-free after TIL treatment. These results demonstrate that translation of meloe transcript in melanoma cells can produce at least two immunogenic polypeptides, MELOE-1 and MELOE-2, from two distinct ORFs that could be relevant target for melanoma immunotherapy.
Collapse
Affiliation(s)
- Yann Godet
- CRCNA, Institut de Recherche Therapeutique, Université de Nantes, UMR INSERM U892, 8 Quai Moncousu, BP70721, 44007 Nantes cedex 1, France
| | - Agnès Moreau-Aubry
- CRCNA, Institut de Recherche Therapeutique, Université de Nantes, UMR INSERM U892, 8 Quai Moncousu, BP70721, 44007 Nantes cedex 1, France
- Faculté des Sciences, Université de Nantes, 44322 Nantes, France
| | - Dimitri Mompelat
- CRCNA, Institut de Recherche Therapeutique, Université de Nantes, UMR INSERM U892, 8 Quai Moncousu, BP70721, 44007 Nantes cedex 1, France
| | - Virginie Vignard
- CRCNA, Institut de Recherche Therapeutique, Université de Nantes, UMR INSERM U892, 8 Quai Moncousu, BP70721, 44007 Nantes cedex 1, France
| | - Amir Khammari
- CRCNA, Institut de Recherche Therapeutique, Université de Nantes, UMR INSERM U892, 8 Quai Moncousu, BP70721, 44007 Nantes cedex 1, France
- Unit of Skin Cancer, Centre Hospitalo-Universitaire de Nantes, 44093 Nantes, France
| | - Brigitte Dreno
- CRCNA, Institut de Recherche Therapeutique, Université de Nantes, UMR INSERM U892, 8 Quai Moncousu, BP70721, 44007 Nantes cedex 1, France
- Unit of Skin Cancer, Centre Hospitalo-Universitaire de Nantes, 44093 Nantes, France
| | - Francois Lang
- CRCNA, Institut de Recherche Therapeutique, Université de Nantes, UMR INSERM U892, 8 Quai Moncousu, BP70721, 44007 Nantes cedex 1, France
- Université de Nantes, UFR des Sciences Pharmaceutiques, 44322 Nantes, France
| | - Francine Jotereau
- CRCNA, Institut de Recherche Therapeutique, Université de Nantes, UMR INSERM U892, 8 Quai Moncousu, BP70721, 44007 Nantes cedex 1, France
- Faculté des Sciences, Université de Nantes, 44322 Nantes, France
| | - Nathalie Labarriere
- CRCNA, Institut de Recherche Therapeutique, Université de Nantes, UMR INSERM U892, 8 Quai Moncousu, BP70721, 44007 Nantes cedex 1, France
| |
Collapse
|
52
|
Bouquié R, Bonnin A, Bernardeau K, Khammari A, Dréno B, Jotereau F, Labarrière N, Lang F. A fast and efficient HLA multimer-based sorting procedure that induces little apoptosis to isolate clinical grade human tumor specific T lymphocytes. Cancer Immunol Immunother 2009; 58:553-66. [PMID: 18751701 PMCID: PMC11029847 DOI: 10.1007/s00262-008-0578-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2008] [Accepted: 08/12/2008] [Indexed: 12/01/2022]
Abstract
HLA multimers are now widely used to stain and sort CD8 T lymphocytes specific for epitopes from viral or tumoral antigens presented in an HLA class I context. However, the transfer of this technology to a clinical setting to obtain clinical grade CD8 T lymphocytes that may be used in adoptive cell transfer (ACT) is hindered by two main obstacles: the first obstacle is the use of streptavidin or derived products that are not available in clinical grade to multimerize HLA/peptide monomers and the second is the reported high degree of apoptosis that eventually occurs when T cell receptors are crosslinked by HLA multimers. In the present report, we describe new HLA multimers composed of immunomagnetic beads covalently coupled to a mAb specific for the AviTag peptide and coated with HLA/peptide monomers bearing the non biotinylated AviTag at the COOH terminus of the HLA heavy chain. Thus, all the components of this new reagent can be obtained in clinical grade. We compared these new multimers with the previously described multimers made with streptavidin beads coated with biotinylated HLA/peptide monomers, in terms of sorting efficiency, recovery of functional T cells, apoptosis and activation. We provide evidence that the new multimers could very efficiently sort pure populations of T lymphocytes specific for three different melanoma antigens (Melan-A, gp100 and NA17-A) after a single peptide stimulation of melanoma patients' PBMC. The recovered specific T cells were cytotoxic against the relevant melanoma cell-lines and, in most cases, produced cytokines. In addition, in marked contrast with streptavidin-based multimers, our new multimers induced very little apoptosis or activation after binding specific T lymphocytes. Altogether, these new multimers fulfill all the necessary requirements to select clinical grade T lymphocytes and should facilitate the development of ACT protocols in cancer patients.
Collapse
Affiliation(s)
- Régis Bouquié
- INSERM U892, 9 quai Moncousu, 44093 Nantes Cedex 1, France
| | | | | | - Amir Khammari
- CHU of Nantes, Unit of Skin Cancer, 44093 Nantes, France
| | - Brigitte Dréno
- INSERM U892, 9 quai Moncousu, 44093 Nantes Cedex 1, France
- CHU of Nantes, Unit of Skin Cancer, 44093 Nantes, France
| | - Francine Jotereau
- INSERM U892, 9 quai Moncousu, 44093 Nantes Cedex 1, France
- Faculté des Sciences, Université de Nantes, 44322 Nantes, France
| | | | - François Lang
- INSERM U892, 9 quai Moncousu, 44093 Nantes Cedex 1, France
- UFR des Sciences Pharmaceutiques, Université de Nantes, 44322 Nantes, France
| |
Collapse
|
53
|
Abstract
The immune response to cancer has been long recognized, including both innate and adaptive responses, showing that the immune system can recognize protein products of genetic and epigenetic changes in transformed cells. The accumulation of antigen-specific T cells within the tumor, the draining lymph node, and the circulation, either in newly diagnosed patients or resultant from experimental immunotherapy, proves that tumors produce antigens and that priming occurs. Unfortunately, just as obviously, tumors grow, implying that anti-tumor immune responses are either not sufficiently vigorous to eliminate the cancer or that anti-tumor immunity is suppressed. Both possibilities are supported by current data. In experimental animal models of cancer and also in patients, systemic immunity is usually not dramatically suppressed, because tumor-bearing animals and patients develop T-cell-dependent immune responses to microbes and to either model antigens or experimental cancer vaccines. However, inhibition of specific anti-tumor immunity is common, and several possible explanations of tolerance to tumor antigens or tumor-induced immunesuppression have been proposed. Inhibition of effective anti-tumor immunity results from the tumor or the host response to tumor growth, inhibiting the activation, differentiation, or function of anti-tumor immune cells. As a consequence, anti-tumor T cells cannot respond productively to developmental, targeting, or activation cues. While able to enhance the number and phenotype of anti-tumor T cells, the modest success of immunotherapy has shown the necessity to attempt to reverse tolerance in anti-tumor T cells, and the vanguard of experimental therapy now focuses on vaccination in combination with blockade of immunosuppressive mechanisms. This review discusses several potential mechanisms by which anti-tumor T cells may be inhibited in function.
Collapse
Affiliation(s)
- Alan B Frey
- Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA.
| | | |
Collapse
|
54
|
Wang RF. Molecular cloning and characterization of MHC class I- and II-restricted tumor antigens recognized by T cells. CURRENT PROTOCOLS IN IMMUNOLOGY 2009; Chapter 20:20.10.1-20.10.29. [PMID: 19235768 DOI: 10.1002/0471142735.im2010s84] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
T cells play a central role in cancer immunosurveillance, autoimmune, and infectious diseases. Identification of MHC class I- and II-restricted T cell peptides is a critical step for the development of effective vaccines against cancer and infectious diseases. This unit describes a cDNA expression system and a genetic targeting expression system for the cloning of genes encoding for MHC class I- and II-restricted antigens recognized by antigen-specific CD8(+) and CD4(+) T cells.
Collapse
Affiliation(s)
- Rong-Fu Wang
- The Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
55
|
Godet Y, Bonnin A, Guilloux Y, Vignard V, Schadendorf D, Dreno B, Jotereau F, Labarriere N. A new tyrosinase epitope recognized in the HLA-B*4002 context by CTL from melanoma patients. Cancer Immunol Immunother 2009; 58:271-80. [PMID: 18612636 PMCID: PMC11031056 DOI: 10.1007/s00262-008-0551-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2007] [Accepted: 03/13/2008] [Indexed: 11/29/2022]
Abstract
Melanoma reactive CTL were obtained by stimulating PBL from a melanoma patient in remission since 1994 following adjuvant TIL immunotherapy, with the autologous melanoma cell line. They were cloned by limiting dilution. One CTL clone recognized melanoma cell lines expressing tyrosinase and the B*4002 molecule, either spontaneously or upon transfection. We demonstrated that this clone recognizes the tyrosinase-derived nonapeptide 316-324 (ADVEFCLSL) and the overlapping decapeptide 315-324 (SADVEFCLSL). We derived two distinct additional specific CTL clones from this same patient that were also reactive against B*4002 melanoma cell lines, suggesting a relative diversity of this specific repertoire in this patient. Stimulating PBMC derived from four additional B*4002 melanoma patients with the tyrosinase 316-324 nonapeptide induced the growth of specific cells for two of the patients, demonstrating the immunogenicity of this new epitope. Our data show that this nonapeptide is a new tool that could be used to generate melanoma-specific T cells for adoptive immunotherapy or serve as a peptide vaccine for HLA-B*4002 melanoma patients.
Collapse
Affiliation(s)
- Yann Godet
- INSERM U892, 9 quai Moncousu, 44093 Nantes cedex 1, France
| | | | - Yannik Guilloux
- INSERM U892, 9 quai Moncousu, 44093 Nantes cedex 1, France
- Université de Nantes, Faculté des Sciences, 44322 Nantes, France
| | | | - Dirk Schadendorf
- Skin Cancer Unit, German Cancer Research Center and University Hospital Mannheim, 68135 Mannheim, Germany
| | - Brigitte Dreno
- INSERM U892, 9 quai Moncousu, 44093 Nantes cedex 1, France
- CHU of Nantes, Unit of Skin Cancer, 44093 Nantes, France
| | - Francine Jotereau
- INSERM U892, 9 quai Moncousu, 44093 Nantes cedex 1, France
- Université de Nantes, Faculté des Sciences, 44322 Nantes, France
| | | |
Collapse
|
56
|
Godet Y, Moreau-Aubry A, Guilloux Y, Vignard V, Khammari A, Dreno B, Jotereau F, Labarriere N. MELOE-1 is a new antigen overexpressed in melanomas and involved in adoptive T cell transfer efficiency. ACTA ACUST UNITED AC 2008; 205:2673-82. [PMID: 18936238 PMCID: PMC2571940 DOI: 10.1084/jem.20081356] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
A cytotoxic T lymphocyte (CTL) clone was derived from a tumor-infiltrating lymphocyte (TIL) population infused to a melanoma patient who remained relapse free for 10 yr after this adoptive transfer. This clone recognized all melanoma cell lines tested and, to a lower extent, melanocytes, in the context of human histocompatibility leukocyte antigen A2 (HLA-A2), but it did not recognize other tumor cell types. The gene coding for the antigen recognized by this clone was identified by the screening of a melanoma complementary DNA expression library. This antigen is overexpressed in melanomas, compared with other cancer cell lines and healthy tissues, and was thus called melanoma-overexpressed antigen (meloe). Remarkably, the structure of meloe was unusual, with multiple short open reading frames (ORFs). The peptide recognized by the CTL clone was encoded by one of these ORFs, called MELOE-1. Using a specific HLA-A2/peptide tetramer, we showed a correlation between the infusion of TILs containing MELOE-1–specific T cells and relapse prevention in HLA-A2 patients. Indeed, 5 out of 9 patients who did not relapse were infused with TILs that contained MELOE-1–specific T cells, whereas 0 out of the 21 patients who relapsed was infused with such TIL-containing lymphocytes. Overall, our results suggest that this new antigen is involved in immunosurveillance and, thus, represents an attractive target for immunotherapy protocols of melanoma.
Collapse
Affiliation(s)
- Yann Godet
- Institut National de Santé et de Recherche Médicale, Unité Mixte de Recherche 892, 44093 Nantes, France
| | | | | | | | | | | | | | | |
Collapse
|
57
|
Carrasco J, Van Pel A, Neyns B, Lethé B, Brasseur F, Renkvist N, van der Bruggen P, van Baren N, Paulus R, Thielemans K, Boon T, Godelaine D. Vaccination of a melanoma patient with mature dendritic cells pulsed with MAGE-3 peptides triggers the activity of nonvaccine anti-tumor cells. THE JOURNAL OF IMMUNOLOGY 2008; 180:3585-93. [PMID: 18292586 DOI: 10.4049/jimmunol.180.5.3585] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
We previously characterized the CTL response of a melanoma patient who experienced tumor regression following vaccination with an ALVAC virus coding for a MAGE-A3 Ag. Whereas anti-vaccine CTL were rare in the blood and inside metastases of this patient, anti-tumor CTL recognizing other tumor Ags, mainly MAGE-C2, were 100 times more frequent in the blood and considerably enriched in metastases following vaccination. In this study we report the analysis of the CTL response of a second melanoma patient who showed a mixed tumor response after vaccination with dendritic cells pulsed with two MAGE-A3 antigenic peptides presented, respectively, by HLA-A1 and HLA-DP4. Anti-MAGE-3.A1 CD8 and anti-MAGE-3.DP4 CD4 T cells became detectable in the blood after vaccination at a frequency of approximately 10(-5) among the CD8 or CD4 T cells, respectively, and they were slightly enriched in slowly progressing metastases. Additional anti-tumor CTL were present in the blood at a frequency of 2x10(-4) among the CD8 T cells and, among these, an anti-MAGE-C2 CTL clone was detected only following vaccination and was enriched by >1,000-fold in metastases relative to the blood. The striking similarity of these results with our previous observations further supports the hypothesis that the induction of a few anti-vaccine T cells may prime or restimulate additional anti-tumor T cell clones that are mainly responsible for the tumor regression.
Collapse
Affiliation(s)
- Javier Carrasco
- Ludwig Institute for Cancer Research, Cellular Genetics Unit, Université Catholique de Louvain, Brussels, Belgium
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
58
|
Hersey P, Halliday GM, Farrelly ML, DeSilva C, Lett M, Menzies SW. Phase I/II study of treatment with matured dendritic cells with or without low dose IL-2 in patients with disseminated melanoma. Cancer Immunol Immunother 2008; 57:1039-51. [PMID: 18157724 PMCID: PMC11030839 DOI: 10.1007/s00262-007-0435-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2007] [Accepted: 12/02/2007] [Indexed: 02/06/2023]
Abstract
BACKGROUND In the present study, we have examined whether treatment of patients with metastatic melanoma with matured dendritic cell (DC) vaccines with or without low dose IL-2 may improve treatment outcomes. METHODS Sixteen patients received DC vaccines (DCs) sensitized with autologous melanoma lysates and 18 patients received DCs sensitized with peptides from gp100, MART-1, tyrosinase, MAGE-3.A2, MAGE-A10 and NA17. IL-2 was given subcutaneously (sc) at 1 MU/m2 on the second day after each injection for 5-14 days in half of each group. DCs were given by intranodal injection. RESULTS There were 2 partial responses (PR) and 3 with stable disease (SD) in the nine patients receiving DCs + peptides + IL-2, and 1 PR and 1 SD in nine patients treated with DCs + peptides without IL-2. There were only two patients with SD in the group receiving DCs + autologous lysates and no IL-2. Median overall survival for all patients was very good at 18.5 months but this was most probably due to selection of a favourable group of patients for the study. There was no significant difference in survival between the groups by log rank analysis. Treatment was not associated with significant side effects. The quality and yield of the DCs in the preparations were generally good. CONCLUSIONS We conclude that mature DC preparations may be superior to immature DC preparations for presentation of melanoma peptides and that IL-2 may increase clinical responses to the DCs plus peptides. However, in our view the low response rates do not justify the cost and complexity of this treatment approach.
Collapse
Affiliation(s)
- P Hersey
- Oncology and Immunology Unit, Room 443, David Maddison Clinical Sciences Building, Cnr. King & Watt Streets, Newcastle, NSW 2300, Australia.
| | | | | | | | | | | |
Collapse
|
59
|
Elluru SR, Duong van Huyen JP, Delignat S, Kazatchkine MD, Friboulet A, Kaveri SV, Bayry J. Induction of maturation and activation of human dendritic cells: a mechanism underlying the beneficial effect of Viscum album as complimentary therapy in cancer. BMC Cancer 2008; 8:161. [PMID: 18533025 PMCID: PMC2442603 DOI: 10.1186/1471-2407-8-161] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2007] [Accepted: 06/04/2008] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Viscum album (VA) preparations have been used as a complimentary therapy in cancer. In addition to their cytotoxic properties, they have also been shown to have immunostimulatory properties. In the present study, we examine the hypothesis that the VA preparations induce activation of human DC that facilitates effective tumor regression. METHODS Four day old monocyte-derived immature DCs were treated with VA Qu Spez at 5, 10 and 15 microg/ml for 48 hrs. The expression of surface molecules was analyzed by flow cytometry. The ability of Qu Spez-educated DC to stimulate T cells was analyzed by allogeneic mixed lymphocyte reaction and activation of Melan-A/MART-1-specific M77-80 CD8+T cells. Cytokines in cell free culture supernatant was analyzed by cytokine bead array assay. RESULTS VA Qu Spez stimulated DCs presented with increased expression of antigen presenting molecule HLA-DR and of co-stimulatory molecules CD40, CD80 and CD86. The VA Qu Spez also induced the secretion of inflammatory cytokines IL-6 and IL-8. Further, Qu Spez-educated DC stimulated CD4+T cells in a allogeneic mixed lymphocyte reaction and activated melanoma antigen Melan-A/MART-1-specific M77-80 CD8+T cells as evidenced by increased secretion of TNF-alpha and IFNgamma. CONCLUSION The VA preparations stimulate the maturation and activation of human DCs, which may facilitate anti-tumoral immune responses. These results should assist in understanding the immunostimulatory properties of VA preparations and improving the therapeutic strategies.
Collapse
Affiliation(s)
- Sri Ramulu Elluru
- Centre de Recherche des Cordeliers, Université Pierre et Marie Curie - Paris6, UMR S 872, Paris, 75006, France.
| | | | | | | | | | | | | |
Collapse
|
60
|
Romero P. Current State of Vaccine Therapies in Non–Small-Cell Lung Cancer. Clin Lung Cancer 2008; 9 Suppl 1:S28-36. [DOI: 10.3816/clc.2008.s.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
61
|
|
62
|
Parmiani G, Russo V, Marrari A, Cutolo G, Casati C, Pilla L, Maccalli C, Rivoltini L, Castelli C. Universal and stemness-related tumor antigens: potential use in cancer immunotherapy. Clin Cancer Res 2007; 13:5675-9. [PMID: 17908956 DOI: 10.1158/1078-0432.ccr-07-0879] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Giorgio Parmiani
- Department of Oncology, San Raffaele Scientific Institute, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
63
|
Levitsky V. Making the tumor-specific effectors ineffective. Semin Cancer Biol 2007; 17:265-6. [PMID: 17624803 DOI: 10.1016/j.semcancer.2007.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
64
|
Benlalam H, Vignard V, Khammari A, Bonnin A, Godet Y, Pandolfino MC, Jotereau F, Dreno B, Labarrière N. Infusion of Melan-A/Mart-1 specific tumor-infiltrating lymphocytes enhanced relapse-free survival of melanoma patients. Cancer Immunol Immunother 2007; 56:515-26. [PMID: 16874485 PMCID: PMC11030260 DOI: 10.1007/s00262-006-0204-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2006] [Accepted: 06/24/2006] [Indexed: 01/25/2023]
Abstract
Adoptive therapy of cancer has been mostly tested in advanced cancer patients using tumor-infiltrating lymphocytes (TIL). Following discouraging results likely due to poor tumor-specificity of TIL and/or high tumor burden, recent studies reiterate the enormous potential of this therapy, particularly in melanoma. We had performed a phase II/III randomised trial on 88 stage III melanoma patients, who received autologous TIL plus IL-2 or IL-2 alone, after complete tumour resection. We reported previously clinical and immunological results supporting the ability of tumor reactive TIL infusion to prevent further development of the melanoma disease and to increase overall survival of patients bearing only one tumor invaded lymph node. The absence of correlation between overall and disease-free survival and the amount of infused tumor-specific TIL suggested that therapeutic efficiency might depend on other parameters such as antigen specificity, function or persistence of TIL. Here we studied the recognition of a panel of 38 shared tumor-associated antigens (TAA) by TIL infused to the patients included in this assay, in order to determine if treatment outcome could correlate with particular antigen specificities of infused TIL. Results show that the infusion of Melan-A/MART-1 reactive TIL appears to be associated with a longer relapse-free survival for HLA-A2 patients. These results further support the relevance of Melan-A/MART-1 antigen as a prime target for immunotherapy protocols in melanoma.
Collapse
Affiliation(s)
| | | | - Amir Khammari
- INSERM U601, 9 quai Moncousu, 44093 Nantes Cedex 1, France
- CHU of Nantes, Unit of Skin Cancer, 44093 Nantes, France
| | | | - Yann Godet
- INSERM U601, 9 quai Moncousu, 44093 Nantes Cedex 1, France
| | | | - Francine Jotereau
- INSERM U601, 9 quai Moncousu, 44093 Nantes Cedex 1, France
- Faculté des Sciences, Université de Nantes, 44322 Nantes, France
| | - Brigitte Dreno
- INSERM U601, 9 quai Moncousu, 44093 Nantes Cedex 1, France
- CHU of Nantes, Unit of Skin Cancer, 44093 Nantes, France
- CHU of Nantes, Unit of cell and gene therapy, 44093 Nantes, France
| | | |
Collapse
|
65
|
Zhao C, Zhang H, Luan F, Zhang R, Liu M, Hu Z, Fan B. QSAR method for prediction of protein-peptide binding affinity: application to MHC class I molecule HLA-A*0201. J Mol Graph Model 2006; 26:246-54. [PMID: 17275373 DOI: 10.1016/j.jmgm.2006.12.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2006] [Accepted: 12/05/2006] [Indexed: 11/29/2022]
Abstract
The support vector machine (SVM), which is a novel algorithm from the machine learning community, was used to develop quantitative structure-activity relationship (QSAR) models for predicting the binding affinity of 152 nonapeptides, which can bind to class I MHC HLA-A*201 molecule. Each peptide was represented by a large pool of descriptors including constitutional, topological descriptors and physical-chemical properties. The heuristic method (HM) was then used to search the descriptor space for selecting the proper ones responsible for binding affinity. The four descriptors were obtained to build linear models based on HM and nonlinear models based on SVM method. The best results are found using SVM: root mean-square (RMS) errors for training, test and whole data set were 0.383, 0.385 and 0.384, respectively. This paper allow the prediction of the binding affinity of new, untested peptides and, through the analysis of contribution of each parameter of different residue at specific position of peptidic ligands, to understand nature of the forces governing binding behavior and suggest new ideas for further synthesis of high-affinity peptides.
Collapse
Affiliation(s)
- Chunyan Zhao
- Department of Chemistry, Lanzhou University, Lanzhou 730000, China
| | | | | | | | | | | | | |
Collapse
|
66
|
Xiong Z, Liu E, Yan Y, Silver RT, Yang F, Chen IH, Chen Y, Verstovsek S, Wang H, Prchal J, Yang XF. An unconventional antigen translated by a novel internal ribosome entry site elicits antitumor humoral immune reactions. THE JOURNAL OF IMMUNOLOGY 2006; 177:4907-16. [PMID: 16982933 PMCID: PMC3902139 DOI: 10.4049/jimmunol.177.7.4907] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Self-tumor Ags that elicit antitumor immune responses in responses to IFN-alpha stimulation remain poorly defined. We screened a human testis cDNA library with sera from three polycythemia vera patients who responded to IFN-alpha and identified a novel Ag, MPD6. MPD6 belongs to the group of cryptic Ags without conventional genomic structure and is encoded by a cryptic open reading frame located in the 3'-untranslated region of myotrophin mRNA. MPD6 elicits IgG Ab responses in a subset of polycythemia vera patients, as well as patients with chronic myelogenous leukemia and prostate cancer, suggesting that it is broadly immunogenic. The expression of myotrophin-MPD6 transcripts was up-regulated in some tumor cells, but only slightly increased in K562 cells in response to IFN-alpha treatment. By using bicistronic reporter constructs, we showed that the translation of MPD6 was mediated by a novel internal ribosome entry site (IRES) upstream of the MPD6 reading frame. Furthermore, the MPD6-IRES-mediated translation, but not myotrophin-MPD6 transcription, was significantly up-regulated in response to IFN-alpha stimulation. These findings demonstrate that a novel IRES-mediated mechanism may be responsible for the translation of unconventional self-Ag MPD6 in responsive to IFN-alpha stimulation. The eliciting antitumor immune response against unconventional Ag MPD6 in patients with myeloproliferative diseases suggests MPD6 as a potential target of novel immunotherapy.
Collapse
MESH Headings
- Amino Acid Sequence
- Antigens, Neoplasm/chemistry
- Antigens, Neoplasm/physiology
- Antigens, Neoplasm/ultrastructure
- Blotting, Northern
- Blotting, Western
- Enzyme-Linked Immunosorbent Assay
- Gene Library
- Humans
- Intercellular Signaling Peptides and Proteins/genetics
- Intercellular Signaling Peptides and Proteins/immunology
- Interferon-gamma/immunology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology
- Male
- Molecular Sequence Data
- Polycythemia Vera/immunology
- Prostatic Neoplasms/immunology
- Protein Biosynthesis
- Protein Structure, Secondary
- RNA, Messenger/analysis
- Reverse Transcriptase Polymerase Chain Reaction
- Ribosomes/chemistry
- Ribosomes/genetics
- Ribosomes/ultrastructure
- Testis/physiology
Collapse
Affiliation(s)
- Zeyu Xiong
- Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA 19140
| | - Enli Liu
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030
| | - Yan Yan
- Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA 19140
| | - Richard T. Silver
- Department of Medicine, New York Presbyterian-Weill Cornell Medical Center, New York, NY 10021
| | - Fan Yang
- Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA 19140
| | - Irene H. Chen
- Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA 19140
| | - Yangyang Chen
- Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA 19140
| | - Srdan Verstovsek
- Department of Leukemia, University of Texas M. D. Anderson Cancer Center, Houston, TX 77030
| | - Hong Wang
- Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA 19140
| | - Josef Prchal
- Department of Medicine, University of Utah School of Medicine, Salt Lake City, UT 84112
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030
| | - Xiao-Feng Yang
- Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA 19140
- Address correspondence and reprint requests to Dr. Xiao-Feng Yang, Department of Pharmacology, Temple University School of Medicine, 3420 North Broad Street, Philadelphia, PA 19140.
| |
Collapse
|
67
|
Saikali S, Avril T, Collet B, Hamlat A, Bansard JY, Drenou B, Guegan Y, Quillien V. Expression of nine tumour antigens in a series of human glioblastoma multiforme: interest of EGFRvIII, IL-13Ralpha2, gp100 and TRP-2 for immunotherapy. J Neurooncol 2006; 81:139-48. [PMID: 17004103 DOI: 10.1007/s11060-006-9220-3] [Citation(s) in RCA: 111] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2006] [Accepted: 07/12/2006] [Indexed: 01/03/2023]
Abstract
In this study, we investigated the mRNA and protein expression of nine tumour antigens in human glioblastoma multiforme with a view to their possible use in dendritic cell-based immunotherapy. Expression of ALK, EGFRvIII, GALT3, gp100, IL-13Ralpha2, MAGE-A3, NA17-A, TRP-2 and tyrosinase were studied by real-time RT-PCR on frozen tissues using a series of 47 tumour samples from patients with glioblastoma. Results were compared with non-neoplastic brain expression or glioblastoma samples with very low levels of expression near the limits of detection for EGFRvIII and MAGE-A3, as these latter two antigens were not detected in non-neoplastic brain. Tumour antigens showing a 5-fold increase in mRNA expression were considered as positive, and only antigens displaying an mRNA over-expression in a significant number of cases were analysed by immunohistochemistry on paraffin-embedded sections. Using real time RT-PCR, we found EGFRvIII, gp100, IL-13Ralpha2 and TRP-2 to be positive in 64, 38, 32 and 21% of cases, respectively. While we observed no over-expression for ALK, GALT3 and tyrosinase, 3 samples out of 47 were positive for MAGE-3 and 1 sample for NA17-A. More than 25% of tumour cells showed strong protein expression in 13, 34, 85 and 96% of GBM samples for gp100, TRP-2, EGFRvIII and IL-13Ralpha2, respectively. Interestingly, protein expression of at least 3 antigens was observed in 38% of cases. These results point out the importance of EGFRvIII, IL-13Ralpha2 and, to a less extent gp100 and TRP-2, for developing an immunotherapy strategy against glioblastoma.
Collapse
Affiliation(s)
- Stéphan Saikali
- Département d'Anatomie et cytologie pathologiques, Hôpital Pontchaillou, Rennes, France
| | | | | | | | | | | | | | | |
Collapse
|
68
|
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
|
69
|
Trakatelli M, Toungouz M, Blocklet D, Dodoo Y, Gordower L, Laporte M, Vereecken P, Sales F, Mortier L, Mazouz N, Lambermont M, Goldman S, Coulie P, Goldman M, Velu T. A new dendritic cell vaccine generated with interleukin-3 and interferon-beta induces CD8+ T cell responses against NA17-A2 tumor peptide in melanoma patients. Cancer Immunol Immunother 2006; 55:469-74. [PMID: 16133111 PMCID: PMC11030794 DOI: 10.1007/s00262-005-0056-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2005] [Accepted: 07/12/2005] [Indexed: 10/25/2022]
Abstract
Dendritic cells derived from monocytes cultured in the presence of type I interferon were found to induce efficient T cell responses against tumor antigens in vitro. We vaccinated eight stage III or IV melanoma patients with dendritic cells generated with interferon-beta and interleukin-3, activated by poly I: C, and pulsed with the tumor-specific antigen NA17.A2. This dendritic cell vaccine was well-tolerated with only minor and transient flu-like symptoms and inflammatory reactions at the injection sites. In most patients, isotopic imaging documented dendritic cells (DC) migration from the intradermal injection site to the draining lymph nodes. Finally, mixed lymphocyte-peptide culture under limiting dilution conditions followed by tetramer labeling indicated that three out of eight patients mounted a CD8 T cell response against the NA17.A2 antigenic peptide. We conclude that DC generated in type I-IFN represent an interesting alternative to DC generated in IL-4 and GM-CSF for cancer immunotherapy.
Collapse
Affiliation(s)
- Myrto Trakatelli
- Department of Medical Oncology, Erasme Hospital, Université Libre de Bruxelles, Route de Lennik 808, 1070 Brussels, Belgium
- Department of Dermatology, Erasme Hospital, Brussels, Belgium
| | - Michel Toungouz
- Cellular and Molecular Therapy Unit, Erasme Hospital, Brussels, Belgium
| | - Didier Blocklet
- Department of Nuclear Medicine, Erasme Hospital, Brussels, Belgium
| | - Ygierne Dodoo
- Institute of Cellular Pathology, University of Louvain, Brussels, Belgium
| | - Laurence Gordower
- Department of Medical Oncology, Erasme Hospital, Université Libre de Bruxelles, Route de Lennik 808, 1070 Brussels, Belgium
| | | | | | | | | | - Naima Mazouz
- Cellular and Molecular Therapy Unit, Erasme Hospital, Brussels, Belgium
| | | | - Serge Goldman
- Department of Nuclear Medicine, Erasme Hospital, Brussels, Belgium
| | - Pierre Coulie
- Institute of Cellular Pathology, University of Louvain, Brussels, Belgium
| | | | - Thierry Velu
- Department of Medical Oncology, Erasme Hospital, Université Libre de Bruxelles, Route de Lennik 808, 1070 Brussels, Belgium
| |
Collapse
|
70
|
Devêvre E, Romero P, Mahnke YD. LiveCount Assay: concomitant measurement of cytolytic activity and phenotypic characterisation of CD8(+) T-cells by flow cytometry. J Immunol Methods 2006; 311:31-46. [PMID: 16527300 DOI: 10.1016/j.jim.2006.01.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2005] [Revised: 12/26/2005] [Accepted: 01/03/2006] [Indexed: 11/22/2022]
Abstract
Tumour immunologists strive to develop efficient tumour vaccination and adoptive transfer therapies that enlarge the pool of tumour-specific and -reactive effector T-cells in vivo. To assess the efficiency of the various strategies, ex vivo assays are needed for the longitudinal monitoring of the patient's specific immune responses providing both quantitative and qualitative data. In particular, since tumour cell cytolysis is the end goal of tumour immunotherapy, routine immune monitoring protocols need to include a read-out for the cytolytic efficiency of Ag-specific cells. We propose to combine current immune monitoring techniques in a highly sensitive and reproducible multi-parametric flow cytometry based cytotoxicity assay that has been optimised to require low numbers of Ag-specific T-cells. The possibility of re-analysing those T-cells that have undergone lytic activity is illustrated by the concomitant detection of CD107a upregulation on the surface of degranulated T-cells. To date, the LiveCount Assay provides the only possibility of assessing the ex vivo cytolytic activity of low-frequency Ag-specific cytotoxic T-lymphocytes from patient material.
Collapse
Affiliation(s)
- Estelle Devêvre
- Division of Clinical Onco-Immunology, Ludwig Institute for Cancer Research, Lausanne Branch, University Hospital (CHUV), Lausanne, Switzerland
| | | | | |
Collapse
|
71
|
Hodi FS. Well-Defined Melanoma Antigens as Progression Markers for Melanoma: Insights into Differential Expression and Host Response Based on Stage. Clin Cancer Res 2006; 12:673-8. [PMID: 16467076 DOI: 10.1158/1078-0432.ccr-05-2616] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- F Stephen Hodi
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts 02115, USA.
| |
Collapse
|
72
|
Abstract
Every field has its dirty little secrets (DLSs): assumptions based on flimsy evidence, findings that directly contradict prevailing models or so beg comprehension that they cannot even seed reasonable alternative hypotheses. Although our natural tendency is to hug these DLSs, they should be exposed, for it is these gaps in our understanding that point to the path to enlightenment. Here, I discuss some of the DLSs of major histocompatibility complex class I antigen processing.
Collapse
Affiliation(s)
- Jonathan W Yewdell
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, MD 20892-0440, USA.
| |
Collapse
|
73
|
Godefroy E, Moreau-Aubry A, Diez E, Dreno B, Jotereau F, Guilloux Y. alpha v beta3-dependent cross-presentation of matrix metalloproteinase-2 by melanoma cells gives rise to a new tumor antigen. ACTA ACUST UNITED AC 2005; 202:61-72. [PMID: 15998788 PMCID: PMC2212908 DOI: 10.1084/jem.20042138] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
A large array of antigens that are recognized by tumor-specific T cells has been identified and shown to be generated through various processes. We describe a new mechanism underlying T cell recognition of melanoma cells, which involves the generation of a major histocompatibility complex class I–restricted epitope after tumor-mediated uptake and processing of an extracellular protein—a process referred to as cross-presentation—which is believed to be restricted to immune cells. We show that melanoma cells cross-present, in an αvβ3-dependent manner, an antigen derived from secreted matrix metalloproteinase–2 (MMP-2) to human leukocyte antigen A*0201-restricted T cells. Because MMP-2 activity is critical for melanoma progression, the MMP-2 peptide should be cross-presented by most progressing melanomas and represents a unique antigen for vaccine therapy of these tumors.
Collapse
Affiliation(s)
- Emmanuelle Godefroy
- Institut National de la Santé et de la Recherche Médicale, Unité 601, Nantes, France
| | | | | | | | | | | |
Collapse
|
74
|
Kurlender L, Borgono C, Michael IP, Obiezu C, Elliott MB, Yousef GM, Diamandis EP. A survey of alternative transcripts of human tissue kallikrein genes. Biochim Biophys Acta Rev Cancer 2005; 1755:1-14. [PMID: 15878240 DOI: 10.1016/j.bbcan.2005.02.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2004] [Revised: 02/10/2005] [Accepted: 02/11/2005] [Indexed: 12/01/2022]
Abstract
Alternative splicing is prevalent within the human tissue kallikrein gene locus. Aside from being the most important source of protein diversity in eukaryotes, this process plays a significant role in development, physiology and disease. A better understanding of alternative splicing could lead to the use of gene variants as drug targets, therapeutic agents or diagnostic markers. With the rapidly rising number of alternative kallikrein transcripts, classifying new transcripts and piecing together the significance of existing data are becoming increasingly challenging. In this review, we present a systematic analysis of all currently known kallikrein alternative transcripts. By defining a reference form for each of the 15 kallikrein genes (KLK1 to KLK15), we were able to classify alternative splicing patterns. We identified 82 different kallikrein gene transcript forms, including reference forms. Alternative splicing may lead to the synthesis of 56 different protein forms for KLK1-15. In the kallikrein locus, the majority of alternative splicing events occur within the protein-coding region, and to a lesser extent in the 5' untranslated regions (UTRs). The most common alternative splicing event is exon skipping (35%) and the least common events are cryptic exons (3%) and internal exon deletion (3%). Seventy-six percent of kallikrein splice variants that are predicted to encode truncated proteins are the result of frameshifts. Eighty-nine percent of putative proteins encoded by splice variants are predicted to be secreted. Although several reports describe the identification of kallikrein splice variants and their potential clinical utility, this is the first extensive review on this subject. Accumulating evidence suggests that alternative kallikrein forms could be involved in many pathologic conditions or could have practical applications as biomarkers. The organization and analysis of the kallikrein transcripts will facilitate future work in this area and may lead to novel clinical and diagnostic applications.
Collapse
Affiliation(s)
- Lisa Kurlender
- Department of Laboratory Medicine and Pathobiology, University of Toronto, 600 University Avenue, Toronto, Ontario, Canada M5G 1L5
| | | | | | | | | | | | | |
Collapse
|
75
|
Hanada KI, Yang JC. Novel biochemistry: post-translational protein splicing and other lessons from the school of antigen processing. J Mol Med (Berl) 2005; 83:420-8. [PMID: 15759099 DOI: 10.1007/s00109-005-0652-6] [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] [Received: 06/15/2004] [Accepted: 12/17/2004] [Indexed: 10/25/2022]
Abstract
In the past 15 years, the molecular identification of antigens that can mediate the killing of tumor cells by T cells has been vigorously pursued. Molecular identification of tumor-associated antigens not only provided the means to activate or monitor anti-tumor immunity, but also gave insights into new and unexpected biochemical processes that are taking place within cells. Post-translational splicing, a phenomenon previously identified only in lower organisms or plants, has recently been added to the list of atypical processes generating proteins in humans. The proteasome, whose main function is to degrade intracellular proteins, appears to catalyze this splicing reaction. The discovery of post-translational splicing has immediate and important implications for the complexity of the major histocompatibility complex (MHC) class I peptide repertoire and for the immune recognition of self- and foreign peptides.
Collapse
Affiliation(s)
- Ken-ichi Hanada
- Surgery Branch, National Cancer Institute, National Institutes of Health, Clinical Research Center, Bethesda, MD 20892-1201, USA.
| | | |
Collapse
|
76
|
Novellino L, Castelli C, Parmiani G. A listing of human tumor antigens recognized by T cells: March 2004 update. Cancer Immunol Immunother 2005; 54:187-207. [PMID: 15309328 PMCID: PMC11032843 DOI: 10.1007/s00262-004-0560-6] [Citation(s) in RCA: 356] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2004] [Accepted: 04/21/2004] [Indexed: 12/22/2022]
Abstract
The technological advances occurred in the last few years have led to a great increase in the number of tumor associated antigens (TAA) that are currently available for clinical applications. In this review we provide a comprehensive list of human tumor antigens as reported in the literature updated at February 2004. The list includes all T cell-defined epitopes, while excluding analogs or artificially modified epitopes, as well as virus-encoded and antibodies-recognized antigens. TAAs are listed in alphabetical order along with the epitope sequence and the HLA allele which restricts recognition by T cells. Data on the tissue distribution of each antigen are also provided together with an extensive bibliography that allows a rapid search for any additional information may be needed on each single antigen or epitope. Overall, the updated list is a database tool for clinicians, scientists and students who have an interest in the field of tumor immunology and immunotherapy.
Collapse
Affiliation(s)
- Luisa Novellino
- Unit of Immunotherapy of Human Tumors, Istituto Nazionale Tumori, Via G. Venezian 1, 20133 Milan, Italy
| | - Chiara Castelli
- Unit of Immunotherapy of Human Tumors, Istituto Nazionale Tumori, Via G. Venezian 1, 20133 Milan, Italy
| | - Giorgio Parmiani
- Unit of Immunotherapy of Human Tumors, Istituto Nazionale Tumori, Via G. Venezian 1, 20133 Milan, Italy
| |
Collapse
|
77
|
Abstract
A number of melanoma vaccines, made from whole melanoma cells or components of melanoma cells, are being tested in Phase II or III trials in patients after surgical removal of high risk primary or regional lymph node metastases, or in those with disseminated melanoma. During the progress of these trials, a number of melanoma antigens and their peptide epitopes that are recognised by human T-cells have been described. These findings and new information about antigen recognition by human T-cells have made it possible to explore the use of peptide epitopes targeted at T-cells as melanoma vaccines. Preliminary results are encouraging and suggest that it may soon be possible to use well defined vaccines, selected on the basis of the antigenic phenotype of the patient's melanoma and their HLA status. Equally exciting advances have been made preparing and using recombinant viral vectors containing genes that code for melanoma antigens. Experimental studies on the use of naked DNA as vaccines are also proceeding. Several fundamental obstacles preventing the effective use of T-cell epitope vaccines remain. These include selection of HLA and tumour antigen loss variants by the immune system, and conditioning of an ineffective immune response by the growing tumour. These aspects suggest that the development of effective vaccine therapy in the future may require a combination of strategies designed to stimulate HLA-restricted and -non-restricted effector cells, and judicious use of cytokines to obtain an effective immune response.
Collapse
Affiliation(s)
- P Hersey
- Oncology & Immunology Unit, Department of Surgery, John Hunter Hospital, Room 443, David Maddison Clinical Sciences Building, Cnr.King & Watt Street, Newcastle, NSW2300, Australia
| |
Collapse
|
78
|
Kalnina Z, Zayakin P, Silina K, Linē A. Alterations of pre-mRNA splicing in cancer. Genes Chromosomes Cancer 2005; 42:342-57. [PMID: 15648050 DOI: 10.1002/gcc.20156] [Citation(s) in RCA: 141] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Recent genomewide analyses of alternative splicing (AS) indicate that up to 70% of human genes may have alternative splice forms, suggesting that AS together with various posttranslational modifications plays a major role in the production of proteome complexity. Splice-site selection under normal physiological conditions is regulated in the developmental stage in a tissue type-specific manner by changing the concentrations and the activity of splicing regulatory proteins. Whereas spliceosomal errors resulting in the production of aberrant transcripts rarely occur in normal cells, they seem to be an intrinsic property of cancer cells. Changes in splice-site selection have been observed in various types of cancer and may affect genes implicated in tumor progression (for example, CD44, MDM2, and FHIT) and in susceptibility to cancer (for example, BRCA1 and APC). Splicing defects can arise from inherited or somatic mutations in cis-acting regulatory elements (splice donor, acceptor and branch sites, and exonic and intronic splicing enhancers and silencers) or variations in the composition, concentration, localization, and activity of regulatory proteins. This may lead to altered efficiency of splice-site recognition, resulting in overexpression or down-regulation of certain splice variants, a switch in splice-site usage, or failure to recognize splice sites correctly, resulting in cancer-specific splice forms. At least in some cases, changes in splicing have been shown to play a functionally significant role in tumorigenesis, either by inactivating tumor suppressors or by gain of function of proteins promoting tumor development. Moreover, cancer-specific splicing events may generate novel epitopes that can be recognized by the host's immune system as cancer specific and may serve as targets for immunotherapy. Thus, the identification of cancer-specific splice forms provides a novel source for the discovery of diagnostic or prognostic biomarkers and tumor antigens suitable as targets for therapeutic intervention.
Collapse
Affiliation(s)
- Zane Kalnina
- Biomedical Research and Study Centre, University of Latvia, Ratsupites St 1, LV-1067 Riga, Latvia
| | | | | | | |
Collapse
|
79
|
Fayolle C, Bauche C, Ladant D, Leclerc C. Bordetella pertussis adenylate cyclase delivers chemically coupled CD8+ T-cell epitopes to dendritic cells and elicits CTL in vivo. Vaccine 2004; 23:604-14. [PMID: 15542180 DOI: 10.1016/j.vaccine.2004.07.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2004] [Accepted: 07/12/2004] [Indexed: 11/22/2022]
Abstract
The adenylate cyclase (CyaA) produced by Bordetella pertussis is able to deliver CD8+ and CD4+ T-cell epitopes genetically grafted within the catalytic domain of the molecule into antigen presenting cells in vivo. We develop now a new approach in which peptides containing CD8+ epitopes are chemically linked to CyaA. We show that CTL responses were induced in mice immunized with CyaA bearing these CD8+ epitopes. Moreover, we demonstrate that the OVA257-264 CD8+ epitope chemically grafted to CyaA is presented to CD8+ T cells by a mechanism requiring (1) proteasome processing, (2) TAP and (3) neosynthesis of MHC class I molecules. Thus, this novel strategy represents a very versatile system as a single CyaA carrier protein could be easily and rapidly coupled to any desired synthetic peptide.
Collapse
Affiliation(s)
- Catherine Fayolle
- Unité de Biologie des Régulations Immunitaires, INSERM E 352, Institut Pasteur, 25 rue du Docteur Roux, 75724 Paris Cedex 15, France
| | | | | | | |
Collapse
|
80
|
Abstract
The MHC molecules present normal as well as disease-related and pathogen-derived peptides to T cells as a way of alerting the immune system of the health status of a cell. Proteomic technologies involving immunoaffinity purification are now extensively used to separate MHC complexes from their peptide cargo, and then the petides are sequenced by tandem mass spectrometry. The identified peptides are tested as vaccine candidates for viral diseases, immunostimulants for treating cancer, and immune-tolerance-inducing agents for autoimmune disorders. One of the challenges in devising novel HLA-peptide-based immunotherapies is to decipher whether a therapeutic window exists between the induction of tumor immunity and the onset of autoimmunity, which can have dangerous sequelae. This review will cover these topics with an overview of the vast possibilities emerging in the field of proteomic analyses of MHC-bound antigens as novel targets for immunotherapy.
Collapse
Affiliation(s)
- Stacy H Shoshan
- Department of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | | |
Collapse
|
81
|
Zajac P, Oertli D, Marti W, Adamina M, Bolli M, Guller U, Noppen C, Padovan E, Schultz-Thater E, Heberer M, Spagnoli G. Phase I/II clinical trial of a nonreplicative vaccinia virus expressing multiple HLA-A0201-restricted tumor-associated epitopes and costimulatory molecules in metastatic melanoma patients. Hum Gene Ther 2004; 14:1497-510. [PMID: 14577912 DOI: 10.1089/104303403322495016] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
We performed a phase I/II clinical trial in metastatic melanoma patients with an ultraviolet (UV)-inactivated nonreplicating recombinant vaccinia virus enabling the expression, from a single construct, of endoplasmic reticulum-targeted HLA-A0201-restricted Melan-A/MART-1(27-35), gp100(280-288), and tyrosinase(1-9) epitopes, together with CD80 and CD86 costimulatory proteins. Corresponding soluble peptides were used to boost responses and granulocyte-macrophage colony-stimulating factor was used as systemic adjuvant. Safety and immunogenicity, as monitored with in vitro-restimulated peripheral blood mononuclear cells by cytotoxic T lymphocyte precursor (CTLp) frequency analysis and tetramer staining, were specifically addressed. Of 20 patients entering the protocol, 2 had to withdraw because of rapidly progressing disease. Immune responses were evaluated in 18 patients (stage III, n = 5; stage IV, n = 13) and increases in specific CTLp frequencies were observed in 15. In 16 patients responsiveness against all 3 antigens could be analyzed: 7 (43%), including all stage III cases, showed evidence of induction of CTLs specific for the three epitopes, and 2 (12%) and 4 (25%), respectively, showed reactivity against two or one tumor-associated antigen. In three stage IV patients no specific CTL reactivity could be induced. Increases in CTLp frequency were detected mostly after viral vaccine injections. However, in a majority of patients final CTLp levels were comparable to initial levels. Tetramer characterization of Melan-A/MART-1(27-35)-specific CTLs during the protocol also suggested preferential expansion after recombinant virus administration. Vector-specific humoral responses, frequently undetectable in stage IV patients, did not appear to prevent tumor-associated antigen-specific CTL induction. Aside from a single occurrence of transient grade 3 leukopenia, no major clinical toxicity was reported. Seventeen of 18 patients completed the 3-month trial (one patient died before the last delayed-type hypersensitivity test). Three displayed regression of individual metastases, seven had stable disease, and progressive disease was observed in seven patients. This is the first report on the administration of a UV-inactivated recombinant vaccinia virus coexpressing five transgenes in cancer patients. The results described here, in terms of safety and immunogenicity, support the use of this reagent in active specific immunotherapy.
Collapse
Affiliation(s)
- P Zajac
- Institute of Surgical Research and Hospital Management, Department of Research, University Hospital of Basel, ZLF, Laboratory 404, Hebelstrasse 20, CH-4031 Basel, Switzerland.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
82
|
Vigneron N, Stroobant V, Chapiro J, Ooms A, Degiovanni G, Morel S, van der Bruggen P, Boon T, Van den Eynde BJ. An antigenic peptide produced by peptide splicing in the proteasome. Science 2004; 304:587-90. [PMID: 15001714 DOI: 10.1126/science.1095522] [Citation(s) in RCA: 251] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
CD8 T lymphocytes recognize peptides of 8 to 10 amino acids presented by class I molecules of the major histocompatibility complex. Here, CD8 T lymphocytes were found to recognize a nonameric peptide on melanoma cells that comprises two noncontiguous segments of melanocytic glycoprotein gp100(PMEL17). The production of this peptide involves the excision of four amino acids and splicing of the fragments. This process was reproduced in vitro by incubating a precursor peptide of 13 amino acids with highly purified proteasomes. Splicing appears to occur by transpeptidation involving an acyl-enzyme intermediate. Our results reveal an unanticipated aspect of the proteasome function of producing antigenic peptides.
Collapse
Affiliation(s)
- Nathalie Vigneron
- Ludwig Institute for Cancer Research and Cellular Genetics Unit, Université de Louvain, B-1200 Brussels, Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|
83
|
Hogan KT, Coppola MA, Gatlin CL, Thompson LW, Shabanowitz J, Hunt DF, Engelhard VH, Ross MM, Slingluff CL. Identification of Novel and Widely Expressed Cancer/Testis Gene Isoforms That Elicit Spontaneous Cytotoxic T-Lymphocyte Reactivity to Melanoma. Cancer Res 2004; 64:1157-63. [PMID: 14871852 DOI: 10.1158/0008-5472.can-03-2209] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Multiple isoforms (TAG-1, TAG-2a, TAG-2b, and TAG-2c) of a novel cancer/testis antigen gene have been identified and are expressed in 84-88% of melanoma cell lines tested. The tumor antigen (TAG) genes are also expressed in K562, a myelogenous leukemia cell line, and they have homology to two chronic myelogenous leukemia-derived clones and a hepatocellular carcinoma clone in the human expressed sequence tags (EST) database, thus indicating that their expression is not restricted to melanomas. In contrast to the fact that many cancer/testis antigens are poorly immunogenic, the TAG-derived peptide, RLSNRLLLR, is recognized by HLA-A3-restricted, melanoma-specific CTLs that were obtained from a melanoma patient with spontaneous reactivity to the peptide. Unlike most cancer/testis antigen genes which are located on the X chromosome, the TAG genes are located on chromosome 5. The genes have the additional unusual features of being coded for in an open reading frame that is initiated by one of three nonstandard initiation codons, and the sequence coding the RLSNRLLLR peptide crosses an exon-exon boundary. The properties of the TAG antigens indicate that they are excellent vaccine candidates for the treatment of melanoma and perhaps other cancers.
Collapse
Affiliation(s)
- Kevin T Hogan
- Department of Surgery, University of Virginia, and Argonex, Inc., Charlottesville, Virginia, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
84
|
Vialle-Castellano A, Laduron S, De Plaen E, Jost E, Dupont S, Ameye G, Michaux L, Coulie P, Olive D, Boon T, van Baren N. A gene expressed exclusively in acute B lymphoblastic leukemias. Genomics 2003; 83:85-94. [PMID: 14667812 DOI: 10.1016/s0888-7543(03)00209-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Representational difference analysis, a cDNA subtraction approach, was used to identify genes that are expressed in acute leukemia but not in normal hematological tissues. We isolated a cDNA fragment from a cell line derived from a B cell acute lymphoblastic leukemia bearing two Philadelphia chromosomes. The cDNA derives from an orphan gene that was named BLACE. BLACE is located in region 7q36 and encodes a major 5.3-kb transcript and several alternatively spliced minor transcripts. Significant expression of BLACE was detected by RT-PCR and quantitative RT-PCR in bone marrow samples from B cell acute lymphoblastic leukemia patients. BLACE was not or was scarcely expressed in other types of hematological malignancies, in normal tissues, and in solid tumors.
Collapse
MESH Headings
- Alternative Splicing
- Blotting, Northern
- Burkitt Lymphoma/genetics
- Burkitt Lymphoma/pathology
- Cell Line, Tumor
- Chromosome Mapping
- Chromosomes, Human, Pair 7/genetics
- DNA, Complementary/chemistry
- DNA, Complementary/genetics
- DNA, Complementary/isolation & purification
- Exons
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic
- Genes/genetics
- Humans
- In Situ Hybridization, Fluorescence
- Introns
- Male
- Middle Aged
- Molecular Sequence Data
- Neoplasm Proteins/genetics
- RNA, Long Noncoding
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Radiation Hybrid Mapping
- Sequence Analysis, DNA
Collapse
Affiliation(s)
- Alexandra Vialle-Castellano
- Laboratoire d'Immunologie des Tumeurs, Institut Paoli-Calmettes, Université de la Méditerranée, Marseille, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
85
|
Gao H, Tian Y, Zhou Y, Chen H, Zhang C, Cheng J, Zhou Y. The polymorphisms of intron1 sequences of HLA-A and -B. Mol Immunol 2003; 40:501-7. [PMID: 14563369 DOI: 10.1016/s0161-5890(03)00224-4] [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: 10/27/2022]
Abstract
Human leucocyte antigen (HLA) class I antigens are highly polymorphic membrane glycoproteins present on most nucleated cells. The polymorphism region is mainly located in exons 2 and 3 of HLA class I genes, which is flanked by introns 1 and 3. Thus, the sequence information of introns 1 and 3 is important for the genotyping of HLA. However, the information about them has not been extensively reported by now. In this work, the intron 1 sequences of HLA-A and -B of 51 standard genomic DNAs provided by the 13th International Histocompatibility Work Group (IHWG) were cloned and sequenced. The polymorphism of the intron sequences were also analyzed by the software Clustal W 1.82. Some sequences were chosen to compare with the standard sequences published in National Center for Biotechnology Information (NCBI). The comparison showed that all the sequences matched exactly with the standard sequences. The knowledge of intron 1 sequences could be very important not only for developing DNA-based typing strategies for the HLA-A and -B alleles but also for establishing an understanding of the evolutionary mechanisms involved in the polymorphism generation of HLA class I alleles.
Collapse
Affiliation(s)
- Huafang Gao
- Department of Biological Sciences and Biotechnology, Tsinghua University, Beijing 100084, PR China
| | | | | | | | | | | | | |
Collapse
|
86
|
Ebeling SB, Ivanov R, Hol S, Aarts TI, Hagenbeek A, Verdonck LF, Petersen EJ. HLA-DRB1*16-restricted recognition of myeloid cells, including CD34+ CML progenitor cells. Br J Haematol 2003; 121:721-9. [PMID: 12780786 DOI: 10.1046/j.1365-2141.2003.04341.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The therapeutic effect of a human leucocyte antigen (HLA)-identical allogeneic stem cell transplantation (allo-SCT) for the treatment of haematological malignancies is mediated partly by the allogeneic T cells that are administered together with the stem cell graft. Chronic myeloid leukaemia (CML) is particularly sensitive to this graft-versus-leukaemia (GVL) effect. Several studies have shown that in allogeneic responses both CD4 and CD8 cells are capable of strong antigen-specific growth inhibition of leukaemic progenitor cells, but that CD4 cells mainly exert the GVL effect against CML. Efficient activation of allogeneic CD4 cells, as well as CD8 cells, may explain the sensitivity of CML cells to elimination by allogeneic T cells. Identification of the antigens recognized by CD4 cells is crucial in understanding the mechanism through which CML cells are so successful in activating allogeneic T cells. In the present report, we describe the characterization of an allogeneic CD4 T-cell clone, DDII.4.4. This clone was found to react against an antigen that is specifically expressed in myeloid cells, including CD34+ CML cells. The antigen recognition is restricted by HLA-DRB1*16. To our knowledge, this is only the second report on an allogeneic CD4 T-cell clone that reacts with early CD34+ myeloid progenitor cells.
Collapse
Affiliation(s)
- Saskia B Ebeling
- Jordan Laboratory for Haemato-Oncology, Department of Haematology, University Medical Centre Utrecht, Utrecht, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
87
|
Uenaka A, Hirano Y, Hata H, Win S, Aji T, Tanaka M, Ono T, Skipper JCA, Shimizu K, Nakayama E. Cryptic CTL epitope on a murine sarcoma Meth A generated by exon extension as a novel mechanism. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 170:4862-8. [PMID: 12707369 DOI: 10.4049/jimmunol.170.9.4862] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Using the recently developed ELISPOT cloning methodology, we obtained cDNA clone S35 coding for the Ag epitope recognized by a murine sarcoma Meth A-specific CTL clone AT-1. Analysis of truncated S35 constructs and overlapping peptides revealed that the peptide epitope was LGAEAIFRL. AT-1 CTL lysed peptide-pulsed CMS8 cells at a nanomolar concentration, and the peptide strongly stimulated IFN-gamma production in AT-1 CTL. Sequence homology indicated that the S35 was derived from a mouse homologue of human retinoic acid-regulated nuclear matrix-associated protein (ramp). The ramp gene consisted of 15 exons. The majority of the ramp mRNA was the transcript normally spliced between exons 14 and 15, but a minor population of mRNA with an extended exon 14 was also present in Meth A cells. The epitope was derived from the newly created open reading frame, which resulted from extension of exon 14 after splicing of the adjacent intronic sequence.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antigens, Neoplasm/biosynthesis
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/isolation & purification
- Antigens, Neoplasm/metabolism
- Base Sequence
- Carrier Proteins/biosynthesis
- Carrier Proteins/genetics
- Carrier Proteins/isolation & purification
- Carrier Proteins/metabolism
- Clone Cells
- Cloning, Molecular
- Cytotoxicity Tests, Immunologic
- DNA-Binding Proteins/biosynthesis
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/isolation & purification
- DNA-Binding Proteins/metabolism
- Enzyme-Linked Immunosorbent Assay
- Epitopes, T-Lymphocyte/biosynthesis
- Epitopes, T-Lymphocyte/genetics
- Epitopes, T-Lymphocyte/isolation & purification
- Epitopes, T-Lymphocyte/metabolism
- Exons/genetics
- Exons/immunology
- Gene Library
- Humans
- Interferon-gamma/analysis
- Methylcholanthrene
- Mice
- Mice, Inbred BALB C
- Mice, Inbred DBA
- Molecular Sequence Data
- Sarcoma, Experimental/chemically induced
- Sarcoma, Experimental/immunology
- T-Lymphocytes, Cytotoxic/immunology
- Transcription Factors
Collapse
Affiliation(s)
- Akiko Uenaka
- Department of Immunology, Okayama University Graduate School of Medicine and Dentistry, Okayama, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
88
|
Nolte A, Scheffold C, Slotty J, Huenefeld C, Schultze JL, Grabbe S, Berdel WE, Kienast J. Generation of melanoma-specific cytotoxic T lymphocytes for allogeneic immunotherapy. J Immunother 2003; 26:257-69. [PMID: 12806279 DOI: 10.1097/00002371-200305000-00009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
To exploit alloreactive T-cell responses known as the graft-versus-tumor effect, allogeneic hematopoietic stem cell transplantation is being explored as experimental therapy in selected solid tumors, including metastatic melanoma. However, donor T-cell responses are often delayed and associated with severe graft-versus-host disease. Posttransplant adoptive immunotherapy using tumor-specific cytotoxic T lymphocytes (CTL) of donor origin might provide immediate graft-versus-tumor effects but not graft-versus-host disease. Therefore, the aim of the current study was to define in vitro conditions for the expansion of allogeneic major histocompatibility complex-matched CTLs targeting melanoma-associated antigens (MAA). The CTLs were generated from peripheral blood mononuclear cells (PBMCs) of HLA-A*0201+ healthy donors by repetitive stimulations with HLA-A*0201-restricted MAA-derived peptides. Melanoma reactivity, as determined by lysis of peptide-pulsed T2 cells and HLA-A2+/Ag+ melanoma cells, was detected using in vitro expanded CTL targeting MAA peptides AAGIGILTV(MT(27-35)), IMDQVPFSV(G(209-2M)), and YMDGTMSQV(T(368-376)). In contrast, FLWGPRALV(MAGE3(271)-(279)) and VLPDVFIRCV(GnT-V(nt38-67)) induced peptide-specific recognition of T2 target cells only, whereas ITDQVPFSV(G(209-217)), KTWGQYWQV(G9(154)), MLLAVLYCL(T(1-9)), and tumor lysate could not induce specific CTLs. Specific cytolytic activity was accompanied by interferon-gamma secretion. Peptide-pulsed dendritic cells were required only for the initial stimulation of CTLs and could be substituted by PBMCs during restimulations. The median expansion rate of CTL was five to six times, regardless of whether dendritic cells or PBMCs were used after the initial stimulation. The results delineate the conditions for effective ex vivo expansion of melanoma-specific CTLs from PBMCs of healthy donors to be used as an adjunct in allogeneic cell therapy of metastatic melanoma.
Collapse
Affiliation(s)
- Annette Nolte
- Department of Medicine/Hematology and Oncology, University of Muenster, Germany
| | | | | | | | | | | | | | | |
Collapse
|
89
|
Marchand M, Punt CJA, Aamdal S, Escudier B, Kruit WHJ, Keilholz U, Håkansson L, van Baren N, Humblet Y, Mulders P, Avril MF, Eggermont AMM, Scheibenbogen C, Uiters J, Wanders J, Delire M, Boon T, Stoter G. Immunisation of metastatic cancer patients with MAGE-3 protein combined with adjuvant SBAS-2: a clinical report. Eur J Cancer 2003; 39:70-7. [PMID: 12504661 DOI: 10.1016/s0959-8049(02)00479-3] [Citation(s) in RCA: 142] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Fifty-seven patients with MAGE-3-positive measurable metastatic cancer, most of them with melanoma, were vaccinated with escalating doses of a recombinant MAGE-3 protein combined with a fixed dose of the immunological adjuvant SBAS-2, which contained MPL and QS21. The immunisation schedule included 4 intramuscular (i.m.) injections at 3-week intervals. Patients whose tumour stabilised or regressed after 4 vaccinations received 2 additional vaccinations at 6-week intervals. The vaccine was generally well tolerated. Among the 33 melanoma patients who were evaluable for tumour response, we observed 2 partial responses, 2 mixed responses and 1 stabilisation. Time to progression in these 5 patients varied from 4 to 29 months. In addition, a partial response lasting 10 months was observed in 1 of the 3 metastatic bladder cancer patients included. None of the tumour responses described above involved visceral metastases. Immunological responses to the vaccine will be reported separately.
Collapse
Affiliation(s)
- M Marchand
- Ludwig Institute for Cancer Research, Brussels Branch, Brussels, Belgium.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
90
|
Schirle M. Identification of tumor-associated HLA-ligands in the post-genomic era. JOURNAL OF HEMATOTHERAPY & STEM CELL RESEARCH 2002; 11:873-81. [PMID: 12590702 DOI: 10.1089/152581602321080538] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Over 10 years ago, the identification of the first tumor-specific T cell epitope shed light on the molecular principles underlying the phenomenon of tumor eradication by the immune system. Since then, a considerable number of different approaches for this task have been introduced and employed successfully, reflecting the growing knowledge about the cellular processes preceding antigen presentation as well as significant technical developments. This review tries to give an overview over available conventional strategies as well as current developments that utilize the potent large-scale screening tools of the post-genomic era.
Collapse
Affiliation(s)
- Markus Schirle
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Germany.
| |
Collapse
|
91
|
Zheng X, Gao JX, Zhang H, Geiger TL, Liu Y, Zheng P. Clonal deletion of simian virus 40 large T antigen-specific T cells in the transgenic adenocarcinoma of mouse prostate mice: an important role for clonal deletion in shaping the repertoire of T cells specific for antigens overexpressed in solid tumors. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 169:4761-9. [PMID: 12391185 DOI: 10.4049/jimmunol.169.9.4761] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
In addition to their overexpression in cancer cells, most of the tumor-associated Ags are expressed at low but detectable levels in normal tissues. It is not clear whether the repertoire of T cells specific for unmutated tumor Ags is shaped by negative selection during T cell development. The transgenic adenocarcinoma of mouse prostate (TRAMP) model is transgenic for the SV40 large T Ag (Tag) under the control of the rat probasin regulatory elements. Although it has been established that T lymphocytes from TRAMP mice are tolerant to SV40 Tag, the mechanism of the tolerance is largely unknown. To examine whether the T cell clonal deletion is responsible for the tolerance, we crossed the TRAMP mice with mice transgenic for a rearranged TCR specific for SV40 Tag presented by the H-2K(k). Double transgenic TRAMP/TCR mice showed profound thymic deletion of SV40 Tag-reactive T cells, including a 6- to 10-fold reduction in the total thymocyte numbers and a >50-fold reduction in phenotypically mature T cells. Consistent with this finding, we observed that the SV40 Tag and endogenous mouse probasin genes are expressed at low levels in the thymus. These results demonstrate that clonal deletion is a major mechanism for tolerance to Ags previously regarded as prostate-specific, and provide direct evidence that the T cell repertoire specific for an unmutated tumor Ag can be shaped by clonal deletion in the thymus.
Collapse
MESH Headings
- Adenocarcinoma/genetics
- Adenocarcinoma/immunology
- Amino Acid Sequence
- Androgen-Binding Protein/biosynthesis
- Animals
- Antigens, Neoplasm/biosynthesis
- Antigens, Neoplasm/genetics
- Antigens, Polyomavirus Transforming/biosynthesis
- Antigens, Polyomavirus Transforming/genetics
- Antigens, Polyomavirus Transforming/immunology
- Cell Line
- Clonal Deletion/genetics
- Epitopes, T-Lymphocyte/biosynthesis
- Epitopes, T-Lymphocyte/genetics
- Epitopes, T-Lymphocyte/immunology
- Female
- Immunodominant Epitopes/immunology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic/genetics
- Mice, Transgenic/immunology
- Molecular Sequence Data
- Prostatic Neoplasms/genetics
- Prostatic Neoplasms/immunology
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- T-Lymphocyte Subsets/virology
- Thymus Gland/cytology
- Thymus Gland/immunology
- Thymus Gland/metabolism
- Thymus Gland/virology
Collapse
Affiliation(s)
- Xincheng Zheng
- Department of Pathology and Comprehensive Cancer Center, Ohio State University, Columbus 43210, USA
| | | | | | | | | | | |
Collapse
|
92
|
Abstract
The characterization of tumor antigens recognized by T lymphocytes has provided the opportunity to immunize cancer patients with well-defined peptides. Differentiation and tumor-specific antigens are expressed in a significant proportion of patients with cancer. Pilot studies have involved many patients with melanoma. No major toxicity has been reported after peptide vaccination. The clinical efficacy of peptide vaccines is limited to a minority of patients and the response rate is less than 20%. Some regressions have been complete and long-lasting. The development of this approach, and that of other methods to deliver tumor antigens, depends on clinical empirism to improve the therapeutic efficacy of the vaccine as well as the accurate understanding of the immune mechanisms involved in vivo.
Collapse
Affiliation(s)
- Jean-Pascal Machiels
- Medical Oncology Unit, Cliniques Universitaires Saint-Luc, Université de Louvain, Brussels, Belgium
| | | | | |
Collapse
|
93
|
Wang RF. Enhancing antitumor immune responses: intracellular peptide delivery and identification of MHC class II-restricted tumor antigens. Immunol Rev 2002; 188:65-80. [PMID: 12445282 DOI: 10.1034/j.1600-065x.2002.18807.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The importance of T-cell-mediated antitumor immunity has been demonstrated in both animal models and human cancer therapy. The identification of major histocompatibility complex (MHC) class I-restricted tumor antigens has generated a resurgence of interest in immunotherapy for cancer. However, recent studies suggest that therapeutic strategies that have mainly focused on the use of CD8+ T cells (and MHC class I-restricted tumor antigens) may not be effective in eliminating cancer cells in patients. Novel strategies have been developed for enhancing T-cell responses against cancer by prolonging antigen presentation of dendritic cells to T cells and the inclusion of MHC class II-restricted tumor antigens. identification of MHC class II-restricted tumor antigens, which are capable of stimulating CD4+ T cells, not only aids our understanding of the host immune responses against cancer antigens, but also provides opportunities for developing effective cancer vaccines.
Collapse
Affiliation(s)
- Rong-Fu Wang
- The Center for Cell and Gene Therapy and Department of Immunology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA.
| |
Collapse
|
94
|
Labarrière N, Bretaudeau L, Gervois N, Bodinier M, Bougras G, Diez E, Lang F, Gregoire M, Jotereau F. Apoptotic body-loaded dendritic cells efficiently cross-prime cytotoxic T lymphocytes specific for NA17-A antigen but not for Melan-A/MART-1 antigen. Int J Cancer 2002; 101:280-6. [PMID: 12209980 DOI: 10.1002/ijc.10605] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
DCs hold promise for cancer immunotherapy due to their functional ambivalence: iDCs internalize antigens, then mDCs trigger naive T-cell activation. However, no consensus has been reached concerning the optimal mode of antigen acquisition for efficient cross-priming of TAA-specific CTLs, and this remains a field of investigation. Here, we used highly purified apobodies derived from an HLA-A*0201-negative melanoma line as a source of tumor antigens for HLA-A*0201 DCs. We compared in vitro mDCs loaded with apobodies to DCs loaded with antigenic peptides, NA17-A(1-9) and Melan-A/MART-1(26-35) A27L analogue, for their capacity to stimulate melanoma antigen-specific T cells from autologous PBLs. Apobody phagocytosis did not induce spontaneous DC maturation, but phagocytic DCs were still responsive to maturation signals, resulting in a functional ability to activate antigen-specific lymphocytes. NA17-A-specific T lymphocytes were activated by both types of stimulation, whereas only peptide-pulsed DCs stimulated the growth of Melan-A/MART-1-specific lymphocytes. We also observed a lack of staining of melanoma-derived apobodies with a Melan-A-specific MAb, suggesting protein alteration during apoptosis induction. After HLA-A*0201/NA17-A multimer sorting, antigen-specific lymphocytes induced by mature DCs loaded with either peptide or apobodies displayed similar functional capacity against peptide-pulsed T2 cells and melanoma cells. Therefore, apobody-loaded DCs can achieve T-cell priming similar to that induced by peptide-pulsed DCs, provided that the apoptotic process allows the preservation of antigen expression.
Collapse
|
95
|
Parmiani G, Castelli C, Dalerba P, Rivoltini L. T cell response to tumor antigens and its therapeutic use in cancer patients. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2002; 495:403-10. [PMID: 11774601 DOI: 10.1007/978-1-4615-0685-0_59] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/23/2023]
Affiliation(s)
- G Parmiani
- Unit of Immunotherapy of Human Cancer, Istituto Nazionale Tumori, Via G. Venezian 1, 20133, Milano, Italy
| | | | | | | |
Collapse
|
96
|
Ghosh MK, Li CL, Fayolle C, Dadaglio G, Murphy A, Lemonnier FA, Roy P, Leclerc C. Induction of HLA-A2-restricted CTL responses by a tubular structure carrying human melanoma epitopes. Vaccine 2002; 20:2463-73. [PMID: 12057601 DOI: 10.1016/s0264-410x(02)00185-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Epitope-based vaccination strategies designed to induce strong tumor-specific CD8(+) T cell responses are being widely considered for cancer immunotherapy. Here, two recombinant tubular structures, NS1-Mela 1 and NS1-Mela 2, carrying, respectively two HLA-A2 epitopes derived from human melanoma antigens were constructed and their capability to induce CTL responses in vivo were studied in HLA-A2 transgenic mice. Strong CTL responses specific for GnT-V/NA 17-A and gp100 (154-162) epitopes were generated in HLA-A2 transgenic mice immunized by the construct NS1-Mela l carrying these two epitopes. The second construct NS1-Mela 2 carrying both Tyrosinase (369-377Da) and Melan-A/Mart-1 (27-35) epitopes induced a weak Tyrosinase-specific CTL response in mice but failed to induce specific CTL responses against the Melan-A/Mart-1 (27-35) epitope in the tested mice. Thus, recombinant tubular structures containing multiple tumoral epitopes may lead to new strategies for the induction of strong tumor-specific CTL responses in cancer patients.
Collapse
Affiliation(s)
- Mrinal K Ghosh
- Division of Geographic Medicine, Department of Medicine, School of Medicine, University of Alabama at Birmingham, 35924, USA
| | | | | | | | | | | | | | | |
Collapse
|
97
|
Parmiani G, Castelli C, Dalerba P, Mortarini R, Rivoltini L, Marincola FM, Anichini A. Cancer immunotherapy with peptide-based vaccines: what have we achieved? Where are we going? J Natl Cancer Inst 2002; 94:805-18. [PMID: 12048268 DOI: 10.1093/jnci/94.11.805] [Citation(s) in RCA: 282] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Many human tumor-associated antigens (TAAs) have recently been identified and molecularly characterized. When bound to major histocompatibility complex molecules, TAA peptides are recognized by T cells. Clinical studies have therefore been initiated to assess the therapeutic potential of active immunization or vaccination with TAA peptides in patients with metastatic cancer. So far, only a limited number of TAA peptides, mostly those recognized by CD8(+) T cells in melanoma patients, have been clinically tested. In some clinical trials, partial or complete tumor regression was observed in approximately 10%-30% of patients. No serious side effects have been reported. The clinical responses, however, were often not associated with a detectable T-cell-specific antitumor immune response when patients' T cells were evaluated in ex vivo assays. In this review, we analyze the available human TAA peptides, the potential immunogenicity (i.e., the ability to trigger a tumor-specific T-cell response) of TAA peptides in vitro and ex vivo, and the potential to construct slightly modified forms of TAA peptides that have increased T-cell stimulatory activity. We discuss the available data from clinical trials of TAA peptide-based vaccination (including those that used dendritic cells to present TAA peptides), identify possible reasons for the limited clinical efficacy of these vaccines, and suggest ways to improve the clinical outcome of TAA peptide-based vaccination for cancer patients.
Collapse
Affiliation(s)
- Giorgio Parmiani
- Unit of Immunotherapy of Human Tumors, Istituto Nazionale per lo Studio e la Cura dei Tumori, Milan, Italy.
| | | | | | | | | | | | | |
Collapse
|
98
|
Shastri N, Schwab S, Serwold T. Producing nature's gene-chips: the generation of peptides for display by MHC class I molecules. Annu Rev Immunol 2002; 20:463-93. [PMID: 11861610 DOI: 10.1146/annurev.immunol.20.100301.064819] [Citation(s) in RCA: 220] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Gene-chips contain thousands of nucleotide sequences that allow simultaneous analysis of the complex mixture of RNAs transcribed in cells. Like these gene-chips, major histocompatibility complex (MHC) class I molecules display a large array of peptides on the cell surface for probing by the CD8(+) T cell repertoire. The peptide mixture represents fragments of most, if not all, intracellular proteins. The antigen processing machinery accomplishes the daunting task of sampling these proteins and cleaving them into the precise set of peptides displayed by MHC I molecules. It has long been believed that antigenic peptides arose as by-products of normal protein turnover. Recent evidence, however, suggests that the primary source of peptides is newly synthesized proteins that arise from conventional as well as cryptic translational reading frames. It is increasingly clear that for many peptides the C-terminus is generated in the cytoplasm, and N-terminal trimming occurs in the endoplasmic reticulum in an MHC I-dependent manner. Nature's gene-chips are thus both parsimonious and elegant.
Collapse
Affiliation(s)
- Nilabh Shastri
- Division of Immunology, Department of Molecular and Cell Biology, University of California, Berkeley, California 94720-3200, USA.
| | | | | |
Collapse
|
99
|
Zendman AJW, Van Kraats AA, Weidle UH, Ruiter DJ, Van Muijen GNP. The XAGE family of cancer/testis-associated genes: alignment and expression profile in normal tissues, melanoma lesions and Ewing's sarcoma. Int J Cancer 2002; 99:361-9. [PMID: 11992404 DOI: 10.1002/ijc.10371] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The existence of XAGE genes was first reported after database homology searches for PAGE-like sequences identified 3 XAGE EST clusters. One of these clusters, XAGE-1, has in later studies been identified as a cancer/testis-associated gene. Here, we report the expression profiles of all 3 reported XAGE genes, as well as several splice variants of XAGE-1, in normal human tissues, Ewing's sarcoma and melanocytic tumors. We also provide the genetic structure of the corresponding genes. Moreover, by searching the databases for XAGE homologues, we identified 3 additional GAGE-like genes. RT-PCR studies showed frequent expression in melanoma metastases and Ewing's sarcoma for 2 XAGE-1-derived transcripts. XAGE-2 was expressed at lower frequency in these tissues, while XAGE-3 was seen only in normal placenta. Due to a frameshift, the largest XAGE-1 putative protein is far less homologous to GAGE-like proteins than the other XAGEs. Interestingly, all GAGE-like genes contain a large secondary open reading frame, coding for putative proteins homologues to the XAGE-1 primary protein. The XAGE family of cancer/testis-associated genes is located on chromosome Xp11.21-Xp11.22. The data outline a superfamily of GAGE-like cancer/testis antigens, consisting of at least 19 genes.
Collapse
Affiliation(s)
- Albert J W Zendman
- Department of Pathology, University Medical Center St. Radboud, Nijmegen, The Netherlands.
| | | | | | | | | |
Collapse
|
100
|
Sahara H, Nabeta Y, Torigoe T, Hirohashi Y, Ichimiya S, Wada Y, Takahashi N, Jimbow K, Yajima T, Watanabe N, Kikuchi K, Sato N. A gene encoding human gastric signet ring cell carcinoma antigen recognized by HLA-A31-restricted cytotoxic T lymphocytes. J Immunother 2002; 25:235-42. [PMID: 12000865 DOI: 10.1097/00002371-200205000-00006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
SUMMARY We previously reported acid-extracted natural antigenic peptide (F4.2 [YSWMDISCWI]) of a gastric signet ring cell carcinoma HST-2 cells, recognized by HLA-A*31012-restricted autologous cytotoxic T lymphocytes, TcHST-2 line. In this study, the full-length cDNA (1101 bp), termed c98, predicting a protein composed of 170 amino acids was obtained. Because TcHST-2 cells could lyse the HLA-A31 antigen (+) allogeneic tumor cells that were introduced with c98 gene, this gene was suggested to possess antigenicity. Beginning at N-terminal 61 amino acid, the N-terminal six amino acid sequence that is completely identical to F4.2 was present in c98; however, a sequence of four amino acids in C-terminal was not found. Nevertheless, this peptide, c98(61-70), seemed to be immunogenic, because cells pulsed with c98(61-70) peptide were lysed in a dose-dependent manner by TcHST-2 cells. The c98 gene was expressed ubiquitously in tumor cells as well as in normal tissues. However, some tumor cells, including HST-2 cells, expressed this antigen in a high content, and such cells were lysed by TcHST-2 cells in the context of HLA-A31 antigen. However, TcHST-2 cells did not lyse cells that expressed lower amounts of c98 than HST-2 cells. These data suggested that c98-gene product and/or c98(61-70) peptides could be used as a candidate for tumor vaccines in cancer immunotherapy.
Collapse
Affiliation(s)
- Hiroeki Sahara
- Marine Biomedical Institute, Sapporo Medical University School of Medicine, Rishirifuji, Hokkaido, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|