101
|
van der Burg SH, de Cock K, Menon AG, Franken KL, Palmen M, Redeker A, Drijfhout J, Kuppen PJ, van de Velde C, Erdile L, Tollenaar RA, Melief CJ, Offringa R. Long lasting p53-specific T cell memory responses in the absence of anti-p53 antibodies in patients with resected primary colorectal cancer. Eur J Immunol 2001; 31:146-55. [PMID: 11169448 DOI: 10.1002/1521-4141(200101)31:1<146::aid-immu146>3.0.co;2-t] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Colorectal carcinoma is commonly associated with mutation and overexpression of p53, making this antigen a potential target for immune intervention. We analyzed humoral and proliferative immunity against p53 in the blood of patients with resected primary colorectal cancer. The majority of these patients displayed anti-p53 T helper (Th) immunity in the absence of measurable p53 specific antibody levels. The Th responses were long-lasting since they could be detected up to several years after resection of the primary tumor. In a number of cases the Th responses were highly sensitive, reflected by the recognition of naturally processed p53 protein. Our data argue that boosting of these responses in patients with minimal residual disease through p53-specific vaccination, may be employed for improving the chance of disease-free survival of these patients.
Collapse
Affiliation(s)
- S H van der Burg
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
102
|
Brunner C, Seiderer J, Schlamp A, Bidlingmaier M, Eigler A, Haimerl W, Lehr HA, Krieg AM, Hartmann G, Endres S. Enhanced dendritic cell maturation by TNF-alpha or cytidine-phosphate-guanosine DNA drives T cell activation in vitro and therapeutic anti-tumor immune responses in vivo. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 165:6278-86. [PMID: 11086063 DOI: 10.4049/jimmunol.165.11.6278] [Citation(s) in RCA: 124] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Dendritic cells (DC) manipulated ex vivo can induce tumor immunity in experimental murine tumor models. To improve DC-based tumor vaccination, we studied whether DC maturation affects the T cell-activating potential in vitro and the induction of tumor immunity in vivo. Maturation of murine bone marrow-derived DC was induced by GM-CSF plus IL-4 alone or by further addition of TNF-alpha or a cytidine-phosphate-guanosine (CpG)-containing oligonucleotide (ODN-1826), which mimics the immunostimulatory effect of bacterial DNA. Flow cytometric analysis of costimulatory molecules and MHC class II showed that DC maturation was stimulated most by ODN-1826, whereas TNF-alpha had an intermediate effect. The extent of maturation correlated with the secretion of IL-12 and the induction of alloreactive T cell proliferation. In BALB/c mice, s.c. injection of colon carcinoma cells resulted in rapidly growing tumors. In this model, CpG-ODN-stimulated DC cocultured with irradiated tumor cells also induced prophylactic protection most effectively and were therapeutically effective when administered 3 days after tumor challenge. Thus, CpG-ODN-enhanced DC maturation may represent an efficient means to improve clinical tumor vaccination.
Collapse
Affiliation(s)
- C Brunner
- Divisions of. Clinical Pharmacology and Neuroendocrinology, Departments of Medicine and Radiation Therapy, Ludwig-Maximilians-University of Munich, Munich, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
103
|
Hoffmann TK, Nakano K, Elder EM, Dworacki G, Finkelstein SD, Appella E, Whiteside TL, DeLeo AB. Generation of T cells specific for the wild-type sequence p53(264-272) peptide in cancer patients: implications for immunoselection of epitope loss variants. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 165:5938-44. [PMID: 11067956 DOI: 10.4049/jimmunol.165.10.5938] [Citation(s) in RCA: 70] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Alterations in the p53 gene occur frequently and can lead to accumulation of p53 protein in squamous cell carcinomas of the head and neck (SCCHN). Since accumulation of p53 is associated with enhanced presentation of wild-type sequence (wt) p53 peptides to immune cells, the development of pan vaccines against SCCHN has focused on wt p53 epitopes. We used the HLA-A2.1-restricted wt p53(264-272) epitope to generate CTL from circulating precursor T cells of HLA-A2.1(+) healthy donors and patients with SCCHN. Autologous peptide-pulsed dendritic cells were used for in vitro sensitization. CTL specific for the wt p53(264-272) peptide were generated from PBMC obtained from two of seven normal donors and three of seven patients with SCCHN. These CTL were HLA class I restricted and responded to T2 cells pulsed with p53(264-272) peptide as well as HLA-A2-matched SCCHN cell lines naturally presenting the epitope. Paradoxically, none of the tumors in the three patients who generated CTL could adequately present the epitope; two had a wt p53 genotype and no p53 protein accumulation, while the third tumor expressed a point mutation (R to H) in codon 273 that prevents presentation of the p53(264-272) epitope. In contrast, patients who did not generate CTL had tumors that accumulated altered p53 and potentially could present the p53(264-272) epitope. These findings suggest that in vivo, CTL specific for the wt p53(264-272) peptide might play a role in the elimination of tumor cells expressing this epitope and in immunoselection of epitope-loss tumor cells. Immunoselection of tumors that become resistant to anti-p53 immune responses has important implications for future p53-based vaccination strategies.
Collapse
MESH Headings
- Autoantibodies/blood
- Carcinoma, Squamous Cell/genetics
- Carcinoma, Squamous Cell/immunology
- Cytotoxicity, Immunologic/genetics
- DNA Mutational Analysis
- Epitopes, T-Lymphocyte/genetics
- Epitopes, T-Lymphocyte/immunology
- Epitopes, T-Lymphocyte/metabolism
- Flow Cytometry
- Genetic Variation/immunology
- HLA-A2 Antigen/immunology
- HLA-A2 Antigen/metabolism
- Head and Neck Neoplasms/genetics
- Head and Neck Neoplasms/immunology
- Humans
- Immunodominant Epitopes/genetics
- Immunodominant Epitopes/immunology
- Immunodominant Epitopes/metabolism
- Lymphocyte Activation/genetics
- Peptide Fragments/genetics
- Peptide Fragments/immunology
- Peptide Fragments/metabolism
- Staining and Labeling
- T-Lymphocyte Subsets/chemistry
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- T-Lymphocytes, Cytotoxic/chemistry
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
- Tumor Cells, Cultured
- Tumor Suppressor Protein p53/biosynthesis
- Tumor Suppressor Protein p53/genetics
- Tumor Suppressor Protein p53/immunology
- Tumor Suppressor Protein p53/metabolism
Collapse
Affiliation(s)
- T K Hoffmann
- University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | | | | | | | | | | | | | | |
Collapse
|
104
|
|
105
|
Abstract
Cytotoxic T Lymphocytes (CTLs) recognize Major Histocompatibility Complex (MHC) class I in complex with peptides. Peptides derived from Tumor Associated Antigens (TAAs) are therefore targets for tumor rejection. A number of TAAs were identified in the last decade from human and murine tumors. Here we summarize the methods for TAA and peptide identification, the nature of TAA peptides and the making of antitumor vaccines.
Collapse
Affiliation(s)
- L Eisenbach
- Department of Immunology, The Weizmann Institute of Science, 76100, Rehovot, Israel.
| | | | | |
Collapse
|
106
|
Schadendorf D, Paschen A, Sun Y. Autologous, allogeneic tumor cells or genetically engineered cells as cancer vaccine against melanoma. Immunol Lett 2000; 74:67-74. [PMID: 10996630 DOI: 10.1016/s0165-2478(00)00251-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Melanoma is a prototype of immunogenic tumor to which various types of immunotherapy have been applied extensively over the past decades. Melanoma vaccines are designed for the purpose of immune modulation and subsequent anti-tumor effects in the process of an active specific immunotherapy. Previous attempts of these vaccines include immunization with whole tumor cells/cell lysates admixed with nonspecific adjuvants. While these vaccines generated enhanced anti-tumor immunity in a subset of patients, some of which showing prolonged survival compared to historical controls, no clinical benefit has so far been demonstrated in a properly controlled phase III study. New-generation melanoma vaccines, which are based on genetic modifications of tumor cells to express cytokines, generated long-lasting systemic anti-tumor immunity in animal models. Translation of these preclinical results primarily into melanoma patients with advanced diseases, shows the potential of these vaccines to induce systemic anti-tumor immune responses and in some instances tumor regression with acceptably low toxicity. Higher efficacy of this novel vaccine approach would be expected when used in a postsurgical adjuvant setting when the tumor load is small. Also other novel vaccine approaches such as dendritic cell-based therapy hold promise for the treatment of melanoma. But the clinical value of all these new approaches has to be analysed in prospectively randomized clinical studies.
Collapse
Affiliation(s)
- D Schadendorf
- Department of Dermatology, Clinical Cooperation Unit for Dermato-Oncology (DKFZ), Clinics Mannheim, University of Heidelberg, Theodor Kutzer Ufer 1, 68135, Mannheim, Germany.
| | | | | |
Collapse
|
107
|
Abstract
The potential to harness the potency and specificity of the immune system underlies the growing interest in cancer immunotherapy. One such approach uses bone marrow-derived dendritic cells, phenotypically distinct and extremely potent antigen-presenting cells, to present tumor-associated antigens and thereby generate tumor-specific immunity. Support for this strategy comes from animal studies that have demonstrated that dendritic cells, when loaded ex vivo with tumor antigens and administered to tumor-bearing hosts, can elicit T cell-mediated tumor destruction. These observations have led to clinical trials designed to investigate the immunologic and clinical effects of antigen-loaded dendritic cells administered as a therapeutic vaccine to patients with cancer. In the design and conduct of such trials, important considerations include antigen selection, methods for introducing the antigen into MHC class I and II processing pathways, methods for isolating and activating dendritic cells, and route of administration. Although current dendritic cell-based vaccination methods are cumbersome, promising results from clinical trials in patients with malignant lymphoma, melanoma, and prostate cancer suggest that immunotherapeutic strategies that take advantage of the antigen presenting properties of dendritic cells may ultimately prove both efficacious and widely applicable to human tumors.
Collapse
Affiliation(s)
- L Fong
- Departments of Pathology and Medicine, Stanford University School of Medicine, Palo Alto, California 94304, USA.
| | | |
Collapse
|
108
|
Dendritic cells modified to express CD40 ligand elicit therapeutic immunity against preexisting murine tumors. Blood 2000. [DOI: 10.1182/blood.v96.1.91] [Citation(s) in RCA: 102] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
CD40 ligand (CD40L) is essential for the initiation of antigen-specific T-cell responses. This study is based on the hypothesis that dendritic cells (DCs) genetically modified ex vivo to express CD40L will enhance in vivo presentation of tumor antigen to the cellular immune system with consequent induction of antitumor immunity to suppress tumor growth. To examine this concept, subcutaneous murine tumors were injected with bone marrow-derived DCs that had been modified in vitro with an adenovirus (Ad) vector expressing murine CD40L (AdmCD40L). In B16 (H-2b, melanoma) and CT26 (H-2d, colon cancer) murine models, intratumoral injection of 2 × 106 AdmCD40L-modified DCs (CD40L-DCs) to established (day 8) subcutaneous tumors resulted in sustained tumor regression and survival advantage. This antitumor effect was sustained when the number of CD40L-DCs were reduced 10-fold to 2 × 105. Analysis of spleens from CD40L-DC–treated animals demonstrated that CD40L-DCs injected into the subcutaneous CT26 flank tumors migrated to the spleen, resulting in activation of immune-relevant processes. Consistent with this concept, intratumoral administration of CD40L-DCs elicited tumor-specific cytotoxic T-lymphocyte responses, and the transfer of spleen cells from CD40L-DC–treated mice efficiently protected naive mice against a subsequent tumor challenge. In a distant 2-tumor model of metastatic disease, an untreated B16 tumor in the right flank regressed in parallel with a left B16 tumor treated with direct injection of CD40L-DCs. These results support the concept that genetic modification of DCs with a recombinant CD40L adenovirus vector may be a useful strategy for directly activating DCs for cancer immunotherapy.
Collapse
|
109
|
Dendritic cells modified to express CD40 ligand elicit therapeutic immunity against preexisting murine tumors. Blood 2000. [DOI: 10.1182/blood.v96.1.91.013k19_91_99] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
CD40 ligand (CD40L) is essential for the initiation of antigen-specific T-cell responses. This study is based on the hypothesis that dendritic cells (DCs) genetically modified ex vivo to express CD40L will enhance in vivo presentation of tumor antigen to the cellular immune system with consequent induction of antitumor immunity to suppress tumor growth. To examine this concept, subcutaneous murine tumors were injected with bone marrow-derived DCs that had been modified in vitro with an adenovirus (Ad) vector expressing murine CD40L (AdmCD40L). In B16 (H-2b, melanoma) and CT26 (H-2d, colon cancer) murine models, intratumoral injection of 2 × 106 AdmCD40L-modified DCs (CD40L-DCs) to established (day 8) subcutaneous tumors resulted in sustained tumor regression and survival advantage. This antitumor effect was sustained when the number of CD40L-DCs were reduced 10-fold to 2 × 105. Analysis of spleens from CD40L-DC–treated animals demonstrated that CD40L-DCs injected into the subcutaneous CT26 flank tumors migrated to the spleen, resulting in activation of immune-relevant processes. Consistent with this concept, intratumoral administration of CD40L-DCs elicited tumor-specific cytotoxic T-lymphocyte responses, and the transfer of spleen cells from CD40L-DC–treated mice efficiently protected naive mice against a subsequent tumor challenge. In a distant 2-tumor model of metastatic disease, an untreated B16 tumor in the right flank regressed in parallel with a left B16 tumor treated with direct injection of CD40L-DCs. These results support the concept that genetic modification of DCs with a recombinant CD40L adenovirus vector may be a useful strategy for directly activating DCs for cancer immunotherapy.
Collapse
|
110
|
Offringa R, Vierboom MP, van der Burg SH, Erdile L, Melief CJ. p53: a potential target antigen for immunotherapy of cancer. Ann N Y Acad Sci 2000; 910:223-33; discussion 233-6. [PMID: 10911916 DOI: 10.1111/j.1749-6632.2000.tb06711.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Approximately 50% of all human malignancies exhibit mutation and aberrant expression of p53, making this protein an interesting candidate target for immunotherapy of cancer. Mutations in p53 are highly diverse. Therefore, targeting of determinants within the wild-type p53 sequence appears most practical. Despite the fact that p53 is ubiquitously expressed, adoptive immunotherapy of tumor-bearing mice with p53-specific cytotoxic T lymphocytes (CTL) results in eradication of p53-overexpressing tumors in the absence of immunopathological damage to normal tissues. These CTL also eliminate tumors that do not show greatly enhanced expression of p53, indicating that the sensitivity of these tumors for p53-specific CTL is determined by the efficiency by which p53-derived peptides are processed into class I MHC, rather than by the steady state levels of p53. Of note, although p53-specific CTL can readily be isolated from p53-/- mice, tolerance for this self antigen may prevent induction of similarly effective CTL in p53+/+ subjects. The T helper (Th) branch of the p53-specific immune response does not seem to be profoundly affected by tolerance. In addition, more and more evidence is obtained for the pivotal role of tumor-specific Th cells in the induction and effector phases of the antitumor response, also against tumors that lack class II MHC expression. The efficacy of Th cells, specific for a recently identified class II MHC-restricted p53 peptide, against p53-overexpressing tumors is currently being investigated. In addition, natural and induced Th responses are analyzed both in a murine tumor model and in a phase I clinical trial involving p53-specific vaccination of colon cancer patients.
Collapse
Affiliation(s)
- R Offringa
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, The Netherlands.
| | | | | | | | | |
Collapse
|
111
|
Fedoseyeva EV, Boisgérault F, Anosova NG, Wollish WS, Arlotta P, Jensen PE, Ono SJ, Benichou G. CD4+ T cell responses to self- and mutated p53 determinants during tumorigenesis in mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 164:5641-51. [PMID: 10820239 DOI: 10.4049/jimmunol.164.11.5641] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We analyzed CD4+ T helper responses to wild-type (wt) and mutated (mut) p53 protein in normal and tumor-bearing mice. In normal mice, we observed that although some self-p53 determinants induced negative selection of p53-reactive CD4+ T cells, other p53 determinants (cryptic) were immunogenic. Next, BALB/c mice were inoculated with J774 syngeneic tumor cell line expressing mut p53. BALB/c tumor-bearing mice mounted potent CD4+ T cell responses to two formerly cryptic peptides on self-p53. This response was characterized by massive production of IL-5, a Th2-type lymphokine. Interestingly, we found that T cell response was induced by different p53 peptides depending upon the stage of cancer. Mut p53 gene was shown to contain a single mutation resulting in the substitution of a tyrosine by a histidine at position 231 of the protein. Two peptides corresponding to wt and mutated sequences of this region were synthesized. Both peptides bound to the MHC class II-presenting molecule (Ed) with similar affinities. However, only mut p53.225-239 induced T cell responses in normal BALB/c mice, a result strongly suggesting that high-affinity wt p53.225-239 autoreactive T cells had been eliminated in these mice. Surprisingly, CD4+ T cell responses to both mut and wt p53.225-239 peptides were recorded in J774 tumor-bearing mice, a phenomenon attributed to the recruitment of low-avidity p53.225-239 self-reactive T cells.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- Cloning, Molecular
- DNA, Complementary/isolation & purification
- Epitopes, T-Lymphocyte/genetics
- Epitopes, T-Lymphocyte/immunology
- Female
- Histocompatibility Antigens Class II/metabolism
- Injections, Intraperitoneal
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Knockout
- Molecular Sequence Data
- Mutation
- Neoplasm Transplantation
- Peptide Fragments/genetics
- Peptide Fragments/immunology
- Sarcoma, Experimental/genetics
- Sarcoma, Experimental/immunology
- Sarcoma, Experimental/metabolism
- Sequence Analysis, DNA
- Tumor Cells, Cultured
- Tumor Suppressor Protein p53/biosynthesis
- Tumor Suppressor Protein p53/genetics
- Tumor Suppressor Protein p53/immunology
- Tumor Suppressor Protein p53/metabolism
Collapse
Affiliation(s)
- E V Fedoseyeva
- Immunogenetics and Transplantation Laboratory, Department of Surgery, University of California, San Francisco, CA 94143, USA
| | | | | | | | | | | | | | | |
Collapse
|
112
|
Fushimi T, Kojima A, Moore MA, Crystal RG. Macrophage inflammatory protein 3alpha transgene attracts dendritic cells to established murine tumors and suppresses tumor growth. J Clin Invest 2000; 105:1383-93. [PMID: 10811846 PMCID: PMC315459 DOI: 10.1172/jci7548] [Citation(s) in RCA: 131] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Dendritic cells (DCs) are powerful antigen-presenting cells that function as the principal activators of T cells. Since the human CC chemokine, macrophage inflammatory protein 3alpha (MIP-3alpha), is chemotactic for DCs in vitro, we hypothesized that adenovirus-mediated gene transfer of MIP-3alpha (AdMIP-3alpha) to tumors might induce local accumulation of DCs and inhibit growth of preexisting tumors. AdMIP-3alpha directed expression of mRNA and protein in vitro, and the supernatant of A549 cells infected with AdMIP-3alpha was chemotactic for DCs. In vivo, injection of AdMIP-3alpha into subcutaneous tumors resulted in local expression of the MIP-3alpha cDNA and in the local accumulation of DCs. In four syngeneic tumor models, growth of established tumors was significantly inhibited compared with untreated tumors or tumors injected with control vector, and in all but the poorly immunogenic LLC carcinoma model, this treatment increased survival advantage of the preexisting tumors. In all four tumor models, intratumoral injection of AdMIP-3alpha induced the local accumulation of CD8b. 2(+) cells and elicited tumor-specific cytotoxic T-lymphocyte activity, and adoptive transfer of splenocytes of animals receiving this treatment protected against a subsequent challenge with the identical tumor cells. In wild-type but not in CD8-deficient mice, AdMIP-3alpha inhibited the growth of tumors. Finally, AdMIP-3alpha also inhibited the growth of distant tumors. This strategy may be useful for enlisting the help of DCs to boost anti-tumor immunity against local and metastatic tumors without the necessity of ex vivo isolation and manipulation of DCs.
Collapse
MESH Headings
- Adenoviridae/genetics
- Adoptive Transfer
- Animals
- Chemokine CCL20
- Chemokines, CC
- Dendritic Cells/immunology
- Dendritic Cells/pathology
- Female
- Genetic Therapy
- Genetic Vectors
- Humans
- Immunity, Cellular
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/pathology
- Macrophage Inflammatory Proteins/genetics
- Macrophage Inflammatory Proteins/immunology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Neoplasms, Experimental/immunology
- Neoplasms, Experimental/pathology
- Neoplasms, Experimental/therapy
- Receptors, CCR6
- Receptors, Chemokine
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/pathology
Collapse
Affiliation(s)
- T Fushimi
- Division of Pulmonary and Critical Care Medicine, Weill Medical College of Cornell University, New York Presbyterian Hospital, New York, New York 10021, USA
| | | | | | | |
Collapse
|
113
|
Linette GP, Shankara S, Longerich S, Yang S, Doll R, Nicolette C, Preffer FI, Roberts BL, Haluska FG. In vitro priming with adenovirus/gp100 antigen-transduced dendritic cells reveals the epitope specificity of HLA-A*0201-restricted CD8+ T cells in patients with melanoma. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 164:3402-12. [PMID: 10706736 DOI: 10.4049/jimmunol.164.6.3402] [Citation(s) in RCA: 47] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Replication-deficient recombinant adenovirus (Ad) encoding human gp100 or MART-1 melanoma Ag was used to transduce human dendritic cells (DC) ex vivo as a model system for cancer vaccine therapy. A second generation E1/E4 region deleted Ad which harbors the CMV immediate-early promoter/enhancer and a unique E4-ORF6/pIX chimeric gene was employed as the backbone vector. We demonstrate that human monocyte-derived DC are permissive to Ad infection at multiplicity of infection between 100 and 500 and occurs independent of the coxsackie Ad receptor. Fluorescent-labeled Ad was used to assess the kinetics and distribution of viral vector within DC. Ad-transduced DC show peak transgene expression at 24-48 h and expression remains detectable for at least 7 days. DC transduced with replication-deficient Ad do not exhibit any unusual phenotypic characteristics or cytopathic effects. DC transduced with Ad2/gp100v2 can elicit tumor-specific CTL in vitro from patients bearing gp100+ metastatic melanoma. Using a panel of gp100-derived synthetic peptides, we show that Ad2/gp100v2-transduced DC elicit Ag-specific CTL that recognize only the G209 and G280 epitopes, both of which display relatively short half-lives ( approximately 7-8 h) on the surface of HLA-A*0201+ cells. Thus, patients with metastatic melanoma are not tolerant to gp100 Ag based on the detection of CD8+ T cells specific for multiple HLA-A*0201-restricted, gp100-derived epitopes.
Collapse
Affiliation(s)
- G P Linette
- Hematology-Oncology Unit and Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
114
|
Perricone MA, Claussen KA, Smith KA, Kaplan JM, Piraino S, Shankara S, Roberts BL. Immunogene therapy for murine melanoma using recombinant adenoviral vectors expressing melanoma-associated antigens. Mol Ther 2000; 1:275-84. [PMID: 10933943 DOI: 10.1006/mthe.2000.0029] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Adenoviral vectors expressing tumor-associated antigens can be used to evoke a specific immune response and inhibit tumor growth. In this study, we tested the efficacy of adenoviral vectors encoding human gp100 (Ad2/hugp100), murine gp100 (Ad2/mugp100), or murine TRP-2 (Ad2/muTRP-2) for their ability to elicit a specific cellular immune response and inhibit the growth of B16 melanoma tumor cells in the mouse. C57BL/6 mice were immunized with Ad2/hugp100, Ad2/mugp100, or Ad2/muTRP-2 either 2 weeks prior to B16-F10 tumor challenge (prophylactic treatment) or 3 days after tumor challenge (active treatment). Ad2/hugp100 and Ad2/muTRP-2 administered to two or more intradermal (i.d.) sites inhibited subsequent subcutaneous tumor growth in > or = 80% of the mice and elicited an antigen-specific cytotoxic T lymphocyte response, whereas other administration routes were not as effective. Ad2/mugp100 administered to two i.d. sites did not inhibit tumor growth or provoke cellular immunity. Immunization was less effective with active treatment where tumor growth was not significantly inhibited by a single dose of either Ad2/muTRP-2 or Ad2/hugp100. However, increasing the number of intradermal immunization sites and the number of doses resulted in progressive improvements in protection from tumor growth in the active treatment model. In conclusion, breaking host tolerance to elicit protective immunity by using adenoviral vectors expressing melanoma-associated antigens is dependent upon the choice of antigen, the site of administration, and the number of doses. These observations provide insights into the clinical applicability of adenoviral vaccines for immunotherapy of malignant diseases.
Collapse
Affiliation(s)
- M A Perricone
- Genzyme Molecular Oncology, Framingham, Massachusetts 01701-9322, USA.
| | | | | | | | | | | | | |
Collapse
|
115
|
Barfoed AM, Petersen TR, Kirkin AF, Thor Straten P, Claesson MH, Zeuthen J. Cytotoxic T-lymphocyte clones, established by stimulation with the HLA-A2 binding p5365-73 wild type peptide loaded on dendritic cells In vitro, specifically recognize and lyse HLA-A2 tumour cells overexpressing the p53 protein. Scand J Immunol 2000; 51:128-33. [PMID: 10652158 DOI: 10.1046/j.1365-3083.2000.00668.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Mutations in the tumour suppressor gene p53 are among the most frequent genetic alterations in human malignancies, often associated with an accumulation of the p53 protein in the cytoplasm. We have generated a number of cytotoxic T lymphocyte (CTL) clones that specifically recognize the HLA-A*0201 p53 wild type peptide RMPEAAPPV [65-73], designated R9V, by the in vitro stimulation of CD8 enriched peripheral blood lymphocytes from a healthy HLA-A*0201 donor using peptide loaded autologous dendritic cells. A total of 22 CTL clones were generated from the same bulk culture and demonstrated to carry identical T-cell receptors. The CTL clone, 2D9, was shown to specifically lyse the HLA-A*0201+ squamous carcinoma cell line SCC9 and the breast cancer cell line MDA-MB-468. Our data demonstrate that human peripheral blood lymphocytes from normal healthy individuals comprise T cells capable of recognizing p53 derived wild type (self) peptides. Furthermore, the capacity of R9V specific T cell clones to exert HLA restricted cytotoxicity, argues that the R9V peptide is naturally presented on certain cancer cells. This supports the view that p53 derived wild type peptides might serve as candidate target antigens for the immunotherapeutic treatment of cancer.
Collapse
Affiliation(s)
- A M Barfoed
- Department of Tumour Cell Biology, Institute of Cancer Biology, Danish Cancer Society, DK-2100 Copenhagen, Denmark
| | | | | | | | | | | |
Collapse
|
116
|
Ribas A, Butterfield LH, Hu B, Dissette VB, Chen AY, Koh A, Amarnani SN, Glaspy JA, McBride WH, Economou JS. Generation of T-cell immunity to a murine melanoma using MART-1-engineered dendritic cells. J Immunother 2000; 23:59-66. [PMID: 10687138 DOI: 10.1097/00002371-200001000-00008] [Citation(s) in RCA: 48] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The murine melanoma B16 expresses the murine counterpart of the human MART-1/Melan-A (MART-1) antigen, sharing a 68.6% amino acid sequence identity. In this study, mice were vaccinated with bone marrow-derived murine dendritic cells genetically modified with a replication-incompetent adenoviral vector to express the human MART-1 gene (AdVMART1). This treatment generated a protective response to a lethal tumor challenge of unmodified murine B16 melanoma cells. The response was mediated by major histocompatibility complex class I-restricted cytotoxic T lymphocytes specific for MART-1 antigen, which produced high levels of interferon-gamma when reexposed to MART-1 in vitro and lysed targets in a calcium-dependent mechanism suggestive of perforin/granzyme B lysis. MART-1 was presented by the dendritic cells used for vaccination and not by epitopes cross-presented by host antigen-presenting cells. In conclusion, dendritic cells genetically modified to express the human MART-1 antigen generate potent murine MART-1-specific protective responses to B16 melanoma.
Collapse
Affiliation(s)
- A Ribas
- Division of Surgical Oncology, University of California, Los Angeles, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
117
|
Tüting T, Steitz J, Brück J, Gambotto A, Steinbrink K, DeLeo AB, Robbins P, Knop J, Enk AH. Dendritic cell-based genetic immunization in mice with a recombinant adenovirus encoding murine TRP2 induces effective anti-melanoma immunity. J Gene Med 1999; 1:400-6. [PMID: 10753065 DOI: 10.1002/(sici)1521-2254(199911/12)1:6<400::aid-jgm68>3.0.co;2-d] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND The induction of cellular immune responses to melanocyte-specific enzymes such as the tyrosinase family of proteins is the goal of various clinical studies for the immunotherapy of melanoma. Tyrosinase-related protein-2 (TRP2) is an attractive model antigen for preclinical studies in C57BL/6 mice because it is naturally expressed by the murine B16 melanoma and can be recognized by self-reactive cytolytic T lymphocytes (CTL). Here we describe efforts to develop genetic immunization with dendritic cells (DC) for the immunotherapy of melanoma in this clinically relevant system. METHODS Recombinant adenoviruses encoding green fluorescent protein (Ad-EGFP) and murine TRP2 (Ad-mTRP2) were constructed using Cre-loxP-mediated recombination. DC were generated in vitro from precursors in bone marrow and transduced with Ad-EGFP or Ad-mTRP2. Mice were immunized by direct injection of adenovirus or by injection of Ad-transduced DC. Induction of tumor immunity was assessed by intravenous challenge with B16 melanoma cells and enumeration of experimentally induced lung metastases. RESULTS Flowcytometric analysis of DC transduced with Ad-EGFP demonstrated endogenous fluorescence due to cytoplasmatic expression of EGFP in 30-60% of cells. Ad-EGFP-transduced DC simultaneously displayed the DC-specific marker NLDC145 and high levels of MHC and costimulatory molecules on their cell surface. Transduction of DC with Ad-mTRP2 resulted in strong intracellular expression of TRP2 which could be readily detected by immunostaining. Importantly, immunization of mice with cultured Ad-mTRP2-transduced DC completely prevented the development of lung metastases following an intravenous challenge with B16 melanoma cells. This striking protective effect was observed with both the intravenous and the subcutaneous route of DC immunization. In vivo depletion of T-cell subsets suggested that the protective effect of an immunization with Ad-mTRP2-transduced DC involved both CD8+ and CD4+ T-cells. CONCLUSIONS Our results demonstrate that DC-based genetic immunization of mice with TRP2, a clinically relevant melanocyte-specific self-antigen, induces effective cellular immunity and prevents metastatic growth of B16 melanoma cells in vivo.
Collapse
Affiliation(s)
- T Tüting
- Department of Dermatology, J. Gutenberg-University, Mainz, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
118
|
PERALTA ELIZABETHA, LIU XIPING, MCCARTY TODDM, WILSON TIMOTHYG, DIAMOND DONJ, ELLENHORN JOSHUAD. IMMUNOTHERAPY OF BLADDER CANCER TARGETING P53. J Urol 1999. [DOI: 10.1016/s0022-5347(05)68242-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Affiliation(s)
- ELIZABETH A. PERALTA
- From the Department of General and Oncologic Surgery, Department of Urology and Urologic Oncology and Department of Hematology Research, City of Hope National Medical Center and Beckman Research Institute, Duarte, California
| | - XIPING LIU
- From the Department of General and Oncologic Surgery, Department of Urology and Urologic Oncology and Department of Hematology Research, City of Hope National Medical Center and Beckman Research Institute, Duarte, California
| | - TODD M. MCCARTY
- From the Department of General and Oncologic Surgery, Department of Urology and Urologic Oncology and Department of Hematology Research, City of Hope National Medical Center and Beckman Research Institute, Duarte, California
| | - TIMOTHY G. WILSON
- From the Department of General and Oncologic Surgery, Department of Urology and Urologic Oncology and Department of Hematology Research, City of Hope National Medical Center and Beckman Research Institute, Duarte, California
| | - DON J. DIAMOND
- From the Department of General and Oncologic Surgery, Department of Urology and Urologic Oncology and Department of Hematology Research, City of Hope National Medical Center and Beckman Research Institute, Duarte, California
| | - JOSHUA D.I. ELLENHORN
- From the Department of General and Oncologic Surgery, Department of Urology and Urologic Oncology and Department of Hematology Research, City of Hope National Medical Center and Beckman Research Institute, Duarte, California
| |
Collapse
|
119
|
|
120
|
Matsue H, Morita A, Matsue K, Takashima A. New technologies toward dendritic cell-based cancer immunotherapies. J Dermatol 1999; 26:757-63. [PMID: 10635619 DOI: 10.1111/j.1346-8138.1999.tb02088.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: 11/30/2022]
Abstract
Immunologically naive T cells are activated most efficiently or even exclusively by special subsets of antigen presenting cells, termed dendritic cells (DC). Members of the DC family have been identified in virtually all epithelial tissues that are constantly exposed to environmental antigens or infectious microbes. For example, skin is equipped with at least two members of this family, epidermal Langerhans cells (LC) and dermal DC. DC have been shown to play pathogenic roles in several different inflammatory/immunological disorders and protective roles against infectious pathogenes and cancer development. In this review article, we will overview the recent progress in the development of DC-based immunotherapies for the prevention and treatment of cancers.
Collapse
Affiliation(s)
- H Matsue
- Department of Dermatology, University of Texas, Southwestern Medical Center, Dallas 75235-9069, USA
| | | | | | | |
Collapse
|
121
|
Affiliation(s)
- E Gilboa
- Department of Surgery, Duke University Medical Center, Durham, North Carolina 27710, USA.
| |
Collapse
|
122
|
Ishida T, Chada S, Stipanov M, Nadaf S, Ciernik FI, Gabrilovich DI, Carbone DP. Dendritic cells transduced with wild-type p53 gene elicit potent anti-tumour immune responses. Clin Exp Immunol 1999; 117:244-51. [PMID: 10444254 PMCID: PMC1905349 DOI: 10.1046/j.1365-2249.1999.00913.x] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
In this study we have tested the concept of using wild-type p53 gene for immunotherapy of cancer. Dendritic cells (DC) were transduced with a human wild-type p53 containing recombinant adenovirus (Ad-p53). About a half of DC transduced with this virus expressed p53 protein by FACS analysis 48 h after infection. Mice immunized twice with Ad-p53 DC developed substantial cytotoxic T lymphocyte (CTL) responses against tumour cells expressing wild-type and different mutant human and murine p53 genes. Very low CTL responses were observed against target cells infected with control adenovirus (Ad-c). Immunization with Ad-p53 provided complete tumour protection in 85% of mice challenged with tumour cells expressing human mutant p53 and in 72.7% of mice challenged with tumour cells with murine mutant p53. Treatment with Ad-p53-transduced DC significantly slowed the growth of established tumours. Thus, DC transduced with wild-type p53 may be a promising new tool for the immunotherapy of cancer.
Collapse
MESH Headings
- Adenoviruses, Human/genetics
- Adenoviruses, Human/immunology
- Animals
- Antineoplastic Agents/immunology
- Antineoplastic Agents/pharmacology
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/genetics
- Cancer Vaccines/immunology
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Dendritic Cells/transplantation
- Dose-Response Relationship, Immunologic
- Female
- Genes, p53/immunology
- Genetic Vectors/administration & dosage
- Genetic Vectors/genetics
- Genetic Vectors/immunology
- Humans
- Immunotherapy, Adoptive/methods
- Injections, Subcutaneous
- Mice
- Mice, Inbred BALB C
- Mice, Inbred CBA
- Neoplasm Transplantation
- Sarcoma, Experimental/genetics
- Sarcoma, Experimental/immunology
- Sarcoma, Experimental/therapy
- Transfection/immunology
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- T Ishida
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | | | | | | | | | | |
Collapse
|
123
|
Affiliation(s)
- P Björck
- Department of Dermatology, BST E1013, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, Pennsylvania, 15261, USA
| |
Collapse
|
124
|
Kaplan JM, Yu Q, Piraino ST, Pennington SE, Shankara S, Woodworth LA, Roberts BL. Induction of Antitumor Immunity with Dendritic Cells Transduced with Adenovirus Vector-Encoding Endogenous Tumor-Associated Antigens. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.163.2.699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
Dendritic cells (DCs) are professional Ag-presenting cells that are being considered as potential immunotherapeutic agents to promote host immune responses against tumor Ags. In this study, recombinant adenovirus (Ad) vectors encoding melanoma-associated Ags were used to transduce murine DCs, which were then tested for their ability to activate CTL and induce protective immunity against B16 melanoma tumor cells. Immunization of C57BL/6 mice with DCs transduced with Ad vector encoding the hugp100 melanoma Ag (Ad2/hugp100) elicited the development of gp100-specific CTLs capable of lysing syngeneic fibroblasts transduced with Ad2/hugp100, as well as B16 cells expressing endogenous murine gp100. The induction of gp100-specific CTLs was associated with long term protection against lethal s.c. challenge with B16 cells. It was also possible to induce effective immunity against a murine melanoma self Ag, tyrosinase-related protein-2, using DCs transduced with Ad vector encoding the Ag. The level of antitumor protection achieved was dependent on the dose of DCs and required CD4+ T cell activity. Importantly, immunization with Ad vector-transduced DCs was not impaired in mice that had been preimmunized against Ad to mimic the immune status of the general human population. Finally, DC-based immunization also afforded partial protection against established B16 tumor cells, and the inhibition of tumor growth was improved by simultaneous immunization against two melanoma-associated Ags as opposed to either one alone. Taken together, these results support the concept of cancer immunotherapy using DCs transduced with Ad vectors encoding tumor-associated Ags.
Collapse
|
125
|
Kerrebijn JD, Balm AJ, Freeman JL, Dosch HM, Drexhage HA. Who is in control of the immune system in head and neck cancer? Crit Rev Oncol Hematol 1999; 31:31-53. [PMID: 10532189 DOI: 10.1016/s1040-8428(99)00011-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Affiliation(s)
- J D Kerrebijn
- Department of Otolaryngology/Head and Neck Surgery, Mount Sinai Hospital, Toronto, Ont., Canada
| | | | | | | | | |
Collapse
|
126
|
Tüting T, Gambotto A, Robbins PD, Storkus WJ, DeLeo AB. Co-delivery of T helper 1-biasing cytokine genes enhances the efficacy of gene gun immunization of mice: studies with the model tumor antigen beta-galactosidase and the BALB/c Meth A p53 tumor-specific antigen. Gene Ther 1999; 6:629-36. [PMID: 10476222 DOI: 10.1038/sj.gt.3300859] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
DNA-based immunization is currently being investigated as a new method for the induction of cellular and humoral immunity directed against viral disease and cancer. In the present study we characterized and compared the immune responses induced in mice following particle-bombardment of the skin ('gene gun' immunization) with those elicited by intracutaneous injection of a recombinant adenoviral vector. Using the well characterized beta-galactosidase (beta gal) model Ag system we find that both in vivo gene transfer systems elicit potent and long-lasting anti-beta gal-specific CD8+ and CD4+ T cell responses. However, gene gun immunization predominantly promotes the production of anti-beta gal antibodies of the gamma 1 isotype, indicative of a Th2-biased immune response, while intradermal injection of recombinant adenovirus primarily leads to the production of anti-beta gal gamma 2a antibodies, indicative of a Th1-biased immune response. Since viral infections are generally associated with the production of large amounts of IFN-alpha and IL-12, we investigated whether administration of expression plasmids encoding these Th1-associated cytokines along with antigen-encoding cDNA can influence the nature of the immune response resulting from gene gun immunization. We observed that co-delivery of IFN-alpha or IL-12 resulted in increased production of anti-beta gal gamma 2a antibodies. This suggests a shift towards a Th1 phenotype of the resulting immune response, thus mimicking a viral infection. Importantly, gene gun immunization of mice with a naturally occurring tumor antigen, the tumor-specific p53 mutant antigen expressed by the chemically induced BALB/c Meth A sarcoma, required co-delivery of IL-12 for the induction of effective antitumor immunity. These results have important implications for the design of clinically relevant gene gun immunization strategies for tumor immunotherapy.
Collapse
Affiliation(s)
- T Tüting
- University of Pittsburgh Cancer Institute, PA 15261, USA
| | | | | | | | | |
Collapse
|
127
|
Men Y, Miconnet I, Valmori D, Rimoldi D, Cerottini JC, Romero P. Assessment of Immunogenicity of Human Melan-A Peptide Analogues in HLA-A*0201/Kb Transgenic Mice. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.162.6.3566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
Previous studies have shown that substitution of single amino acid residues in human Melan-A immunodominant peptides Melan-A27–35 and Melan-A26–35 greatly improved their binding and the stability of peptide/HLA-A*0201 complexes. In particular, one Melan-A peptide analogue was more efficient in the generation of Melan-A peptide-specific and melanoma-reactive CTL than its parental peptide in vitro from human PBL. In this study, we analyzed the in vivo immunogenicity of Melan-A natural peptides and their analogues in HLA-A*0201/Kb transgenic mice. We found that two human Melan-A natural peptides, Melan-A26–35 and Melan-A27–35, were relatively weak immunogens, whereas several Melan-A peptide analogues were potent immunogens for in vivo CTL priming. In addition, induced Melan-A peptide-specific mouse CTL cross-recognized natural Melan-A peptides and their analogues. More interestingly, these mouse CTL were also able to lyse human melanoma cell lines in vitro in a HLA-A*0201-restricted, Melan-A-specific manner. Our results indicate that the HLA-A*0201/Kb transgenic mouse is a useful animal model to perform preclinical testing of potential cancer vaccines, and that Melan-A peptide analogues are attractive candidates for melanoma immunotherapy.
Collapse
Affiliation(s)
- Ying Men
- *Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne, Epalinges, Switzerland; and
| | - Isabelle Miconnet
- *Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne, Epalinges, Switzerland; and
| | - Danila Valmori
- †Division of Clinical Onco-Immunology, Ludwig Institute for Cancer Research, Lausanne Branch, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Donata Rimoldi
- *Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne, Epalinges, Switzerland; and
| | - Jean-Charles Cerottini
- *Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne, Epalinges, Switzerland; and
- †Division of Clinical Onco-Immunology, Ludwig Institute for Cancer Research, Lausanne Branch, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Pedro Romero
- *Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne, Epalinges, Switzerland; and
- †Division of Clinical Onco-Immunology, Ludwig Institute for Cancer Research, Lausanne Branch, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| |
Collapse
|
128
|
Abstract
Human tumors express a number of protein antigens that can be recognized by T cells, thus providing potential targets for cancer immunotherapy. Dendritic cells (DCs) are rare leukocytes that are uniquely potent in their ability to present antigens to T cells, and this property has prompted their recent application to therapeutic cancer vaccines. Isolated DCs loaded with tumor antigen ex vivo and administered as a cellular vaccine have been found to induce protective and therapeutic anti-tumor immunity in experimental animals. In pilot clinical trials of DC vaccination for patients with non-Hodgkin's lymphoma and melanoma, induction of anti-tumor immune responses and tumor regressions have been observed. Additional trials of DC vaccination for a variety of human cancers are under way, and methods for targeting tumor antigens to DCs in vivo are also being explored. Exploitation of the antigen-presenting properties of DCs thus offers promise for the development of effective cancer immunotherapies.
Collapse
Affiliation(s)
- J M Timmerman
- Department of Medicine, Stanford University School of Medicine, California 94305, USA.
| | | |
Collapse
|
129
|
Abstract
Dendritic cells (DC) are potent antigen presenting cells that are essential for the initiation of primary immune responses. They richly express MHC, costimulatory and adhesion molecules necessary for the stimulation of naive T cell populations. Dendritic cells are located at sites of antigen capture where they demonstrate phagocytic capacity and subsequently migrate to lymphatic areas for antigen presentation. Their phenotypic and functional characteristics are intimately linked to their stage of maturation. The hematopoietic development of dendritic cells is distinct and may follow several precursor pathways some closely linked to monocytes. Generation of large numbers of cells for potential clinical use has recently been accomplished through the in vitro culturing of progenitors with cytokines. The use of dendritic cell vaccines for cancer immunotherapy has emerged as an exciting new focus of investigation. Various strategies have been adopted to introduce tumor antigens into dendritic cells so that they may be more effectively presented to T cells in the context of costimulation. Animal models demonstrate that dendritic cell tumor vaccines reverse T-cell anergy and result in subsequent tumor rejection. Incorporating the expanding knowledge of dendritic cell biology into vaccine design is essential for the generation of effective immunotherapy for cancer patients.
Collapse
Affiliation(s)
- D Avigan
- Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215, USA.
| |
Collapse
|
130
|
Tüting T, Wilson CC, Martin DM, Baar J, DeLeo A, Lotze MT, Storkus WJ. DNA vaccines targeting dendritic cells for the immunotherapy of cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 1999; 451:295-304. [PMID: 10026887 DOI: 10.1007/978-1-4615-5357-1_46] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Affiliation(s)
- T Tüting
- Department of Surgery, University of Pittsburgh School of Medicine, PA 15261, USA
| | | | | | | | | | | | | |
Collapse
|
131
|
Abstract
Nearly twenty years after the initial discovery of p53, we are now in an ideal position to exploit our vast knowledge of p53 biology in the creation of novel cancer therapies. Disruption of p53 function through mutation, or other means, occurs very frequently in human cancer. Loss of p53 function has been linked with unfavourable prognosis in a large number of tumour types, as indicated by more aggressive tumours, early metastasis and decreased survival rates. Many different avenues of research have converged upon p53 to highlight this protein as being one of the foremost cellular responders to stress, in particular to DNA damage. Huge advances have been made in understanding the complex role p53 plays in the regulation of apoptosis and cell cycle arrest. This review is not meant to be a comprehensive description of p53 biology, but rather serves to highlight current progress in the development of p53-oriented cancer therapies. These may be categorised into three basic strategies: gene replacement therapy using wild-type p53, restoration of p53 function by other means and, finally, targeting of the p53 dysfunction itself. Rapid progress is expected to be made regarding the identification of conventional pharmaceutical agents which either work in a p53-independent manner or act preferentially in p53 defective cells. Gene replacement therapy with wild-type p53 also holds considerable potential for obtaining clinically relevant results quickly. The other forms of cancer therapies based around p53 are much further behind in the developmental process, but may prove to more efficacious in the long run, especially in terms of specificity. As with many other fields, the innovation of successful p53-oriented cancer therapies is only limited by our understanding of p53 biology and the creative use of such knowledge.
Collapse
Affiliation(s)
- W M Gallagher
- Oncology Department, Rhône-Poulenc Rorer, CRVA, Vitry-sur-Seine, France
| | | |
Collapse
|
132
|
Abstract
Cancer vaccines are a promising tool in the hands of the clinical oncologist. We have summarized the most recent findings and achievements in this exciting field. Tumor-associated antigens, as a basis for the new cancer vaccines, are reviewed. We emphasize novel approaches for the design of safe and more effective vaccines for cancer. We also discuss the possible clinical applications and the future prospects for vaccine development.
Collapse
Affiliation(s)
- B R Minev
- Center for Biological Therapy and Melanoma Research, Cancer Center, University of California-San Diego, La Jolla 92093-0061, USA
| | | | | |
Collapse
|
133
|
De Veerman M, Heirman C, Van Meirvenne S, Devos S, Corthals J, Moser M, Thielemans K. Retrovirally Transduced Bone Marrow-Derived Dendritic Cells Require CD4+ T Cell Help to Elicit Protective and Therapeutic Antitumor Immunity. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.162.1.144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
It has been extensively documented that murine dendritic cells loaded with tumor-associated Ag (TAA)-derived peptides or protein can prime Ag-specific CD8+ cytotoxic T cells in vivo and can elicit Ag-specific immunity. Optimal presentation of TAA might be achieved by retroviral transduction of DCs allowing long term and stable expression of the TAA-peptides as well as the presentation of multiple epitopes in the context of MHC class I and/or class II molecules. Here we show that retroviral transduction of bone marrow-derived dendritic cells (DCs) with chicken OVA cDNA or the reporter gene green fluorescent protein retained their potent stimulatory capacity and that the transduced DCs could process and present the endogenously expressed OVA protein. The DCs transduced with cDNA encoding native OVA protein presented OVA-derived peptides in the context of MHC class I as well as MHC class II and induced a strong Ag-specific CTL response. DCs expressing a cytosolic form of OVA presented OVA peptides only in the context of MHC class I and failed to induce an OVA-specific CTL response in vivo when they had been cultured in the absence of exogenous protein. Immunization with retrovirally transduced DCs resulted in an Ag-specific immunity and rejection of a tumor cell challenge and a significant survival advantage in tumor-bearing mice. These results obtained in this rapidly lethal tumor model suggest that DCs transduced with TAA may be useful for tumor immunotherapy and underscore the importance of the simultaneous delivery of T cell help in the development of Ag-specific cytotoxic T-cells.
Collapse
Affiliation(s)
- Marijke De Veerman
- *Laboratory of Physiology, Medical School of Vrije Universiteit Brussel, Brussels, Belgium; and
| | - Carlo Heirman
- *Laboratory of Physiology, Medical School of Vrije Universiteit Brussel, Brussels, Belgium; and
| | - Sonja Van Meirvenne
- *Laboratory of Physiology, Medical School of Vrije Universiteit Brussel, Brussels, Belgium; and
| | - Sophie Devos
- *Laboratory of Physiology, Medical School of Vrije Universiteit Brussel, Brussels, Belgium; and
| | - Jurgen Corthals
- *Laboratory of Physiology, Medical School of Vrije Universiteit Brussel, Brussels, Belgium; and
| | - Muriel Moser
- †Department of Molecular Biology, Université Libre de Bruxelles, Rhode-St-Genèse, Belgium
| | - Kris Thielemans
- *Laboratory of Physiology, Medical School of Vrije Universiteit Brussel, Brussels, Belgium; and
| |
Collapse
|
134
|
Theobald M, Ruppert T, Kuckelkorn U, Hernandez J, Häussler A, Ferreira EA, Liewer U, Biggs J, Levine AJ, Huber C, Koszinowski UH, Kloetzel PM, Sherman LA. The sequence alteration associated with a mutational hotspot in p53 protects cells from lysis by cytotoxic T lymphocytes specific for a flanking peptide epitope. J Exp Med 1998; 188:1017-28. [PMID: 9743520 PMCID: PMC2212538 DOI: 10.1084/jem.188.6.1017] [Citation(s) in RCA: 90] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/1997] [Revised: 06/25/1998] [Indexed: 12/31/2022] Open
Abstract
A high proportion of tumors arise due to mutation of the p53 tumor suppressor protein. A p53 hotspot mutation at amino acid position 273 from R to H, flanking a peptide epitope that spans residues 264-272, renders cells resistant to killing by human histocompatibility leukocyte antigen (HLA)-A*0201-restricted cytotoxic T lymphocytes (CTLs) specific for this epitope. Acquisition of the R to H mutation at residue 273 of the human p53 protein promotes tumor growth in vivo by selective escape from recognition by p53.264-272 peptide-specific CTLs. Synthetic 27-mer p53 polypeptides covering the antigenic nonamer region 264-272 of p53 were used as proteasome substrates to investigate whether the R to H mutation at the P1' position of the COOH terminus of the epitope affects proteasome-mediated processing of the protein. Analysis of the generated products by tandem mass spectrometry and the kinetics of polypeptide processing in conjunction with CTL assays demonstrate that the R to H mutation alters proteasomal processing of the p53 protein by inhibiting proteolytic cleavage between residues 272 and 273. This prevents the release of the natural CTL epitope that spans flanking residues 264-272 as well as a putative precursor peptide. These results demonstrate that mutation of p53 not only leads to malignant transformation but may also, in some instances, affect immune surveillance and should be considered in the design of cancer vaccines.
Collapse
Affiliation(s)
- M Theobald
- Department of Hematology, Johannes Gutenberg-University, Mainz, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
135
|
Jiao Y, Fujimoto S. Sequential T cell response involved in tumor rejection of sarcoma, Meth A, in syngeneic mice. Jpn J Cancer Res 1998; 89:657-65. [PMID: 9703364 PMCID: PMC5921869 DOI: 10.1111/j.1349-7006.1998.tb03268.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
We investigated the type of T cell response involved in Meth A tumor rejection in primary immune and hyperimmune syngeneic mice. It was found that a CD4+ T cell-mediated delayed-type hypersensitivity (DTH) response activating non-specific killer cells such as macrophages, NK and LAK cells, without a specific CD8+ cytotoxic T lymphocyte (CTL) response, was the major immune response leading to Meth A tumor rejection in primary immune mice. In contrast, the specific CD8+ CTL response was the major response leading to the tumor rejection, in addition to CD4+ T cell-mediated DTH response, in hyperimmune mice. Analysis of CD4+ T cell clones established from primary immune and hyperimmune spleen cells indicated that a CD4+ T cell clone (C9) of primary immune mice (although only one clone was established) was of Th1 type, and induced cytotoxicity in accessory cells by classic DTH in vitro. Eight CD4+ T cell clones were established from hyperimmune spleen cells. Six out of the eight clones were of the Th2 type and two were Th0-like. However, no Th1-type CD4+ T cell clone was established from hyperimmune spleen cells. All of these CD4+ T cell clones, even the Th2-type clones, were capable of inducing cytotoxicity in vitro in T cell-depleted accessory cells, as in an in vitro DTH response. We postulate on the basis of these results that the T cell response leading to Meth A tumor rejection in vivo sequentially changed from a CD4+ T cell-mediated classic DTH response to a CD8+ CTL response, in addition to a cellular response mediated probably by Th2-type cells, during the process of repeated immunization.
Collapse
Affiliation(s)
- Y Jiao
- Department of Immunology, Kochi Medical School
| | | |
Collapse
|
136
|
Toes REM, van der Voort EIH, Schoenberger SP, Drijfhout JW, van Bloois L, Storm G, Kast WM, Offringa R, Melief CJM. Enhancement of Tumor Outgrowth Through CTL Tolerization After Peptide Vaccination Is Avoided by Peptide Presentation on Dendritic Cells. THE JOURNAL OF IMMUNOLOGY 1998. [DOI: 10.4049/jimmunol.160.9.4449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
Synthetic peptide-based vaccines have been shown to induce potent protective and therapeutic T cell-mediated immunity in preclinical animal models and are now being evaluated in clinical phase I/II studies for their efficacy against tumors or infectious diseases. However, such vaccines might also specifically tolerize T cells causing enhanced tumor outgrowth, as shown by vaccination with two CTL epitopes derived from the adenovirus type 5 early region 1 (Ad5E1) oncogenes. We now report that modification of the Ad5E1 peptide vaccine either through incorporation of the peptides into liposomes or by ligation of the peptides to lipid tails, another vaccine formulation being tested in the clinic, fails to convert immunosuppression into effective antitumor vaccination. Inclusion of a helper T cell epitope into the vaccine likewise induces enhanced tumor outgrowth and thus does not diminish the capacity of the peptides to tolerize Ad5E1-specific CTL. In contrast, the Ad5E1-derived peptides evoke a strong tumor-protective CTL response when presented on dendritic cells (DC), indicating that the in vivo CTL-tolerizing potential of these peptides is converted to specific immunostimulation when presented on DC. These findings have important implications for the development of peptide-based immune intervention strategies and emphasize the superior nature of Ag-pulsed DC over other peptide-based vaccination protocols as well as the crucial importance of the mode of peptide-Ag delivery in setting the balance between T cell stimulation and tolerization.
Collapse
Affiliation(s)
- René E. M. Toes
- *Department of Immunohematology and Blood Bank, University of Leiden, Leiden, The Netherlands
| | | | - Stephen P. Schoenberger
- *Department of Immunohematology and Blood Bank, University of Leiden, Leiden, The Netherlands
| | - Jan Wouter Drijfhout
- *Department of Immunohematology and Blood Bank, University of Leiden, Leiden, The Netherlands
| | - Louis van Bloois
- †Department of Pharmaceutics, Faculty of Pharmacy, University of Utrecht, Utrecht, The Netherlands; and
| | - Gert Storm
- †Department of Pharmaceutics, Faculty of Pharmacy, University of Utrecht, Utrecht, The Netherlands; and
| | - W. Martin Kast
- ‡Cancer Immunology Program, Cardinal Bernardin Cancer Center, Loyola University of Chicago, Maywood, IL 60153
| | - Rienk Offringa
- *Department of Immunohematology and Blood Bank, University of Leiden, Leiden, The Netherlands
| | - Cornelis J. M. Melief
- *Department of Immunohematology and Blood Bank, University of Leiden, Leiden, The Netherlands
| |
Collapse
|
137
|
Sionov RV, Haupt Y. Apoptosis by p53: mechanisms, regulation, and clinical implications. SPRINGER SEMINARS IN IMMUNOPATHOLOGY 1998; 19:345-62. [PMID: 9540161 DOI: 10.1007/bf00787230] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- R V Sionov
- Lautenberg Center for General and Tumor Immunology, Hebrew University Hadassah Medical School, Jerusalem, Israel
| | | |
Collapse
|
138
|
Shortt J, Hart DN, Watson JD, Baird MA. Blockade of B7-2, not B7-1, inhibits purified protein derivative-primed T-lymphocyte responses but fails to influence the proportion of Th1 versus Th2 subsets. Scand J Immunol 1998; 47:355-62. [PMID: 9600317 DOI: 10.1046/j.1365-3083.1998.00315.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The ability to select for a cell-mediated response rather than antibody production following infection with intracellular mycobacteria, would be an advantage in preventing the occurrence of disease. Recent work suggests that the two members of the B7 family of costimulatory molecules, B7-1 and B7-2, may differentially influence the nature of primary immune responses but little is known of their role in this capacity in secondary responses. We have used an in vitro model to investigate whether blocking B7-1 and B7-2 affects changes in the cytokine profiles of Th lymphocytes previously primed to purified protein derivative (PPD) from Mycobacterium bovis. In C57BL/6 and BALB/c mice we found that the proliferative responses of a component of recently activated T lymphocytes, and those returning to the resting state, were inhibited by B7-2 blockade. B7-1 blockade had no distinguishable effect. However, in cultures containing anti-B7-2 antibody, the production of both interferon-gamma (IFN-gamma) and interleukin-4 (IL-4), indicative of cell-mediated and antibody responses, respectively, were reduced. This suggests that intervention in a recall response to mycobacterial antigen by blocking B7-1 or B7-2 molecules, is unlikely to alter the nature of the immune response.
Collapse
Affiliation(s)
- J Shortt
- Department of Pathology, Dunedin School of Medicine, New Zealand
| | | | | | | |
Collapse
|
139
|
Nestle FO, Alijagic S, Gilliet M, Sun Y, Grabbe S, Dummer R, Burg G, Schadendorf D. Vaccination of melanoma patients with peptide- or tumor lysate-pulsed dendritic cells. Nat Med 1998; 4:328-32. [PMID: 9500607 DOI: 10.1038/nm0398-328] [Citation(s) in RCA: 2015] [Impact Index Per Article: 74.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Melanoma is the main cause of death in patients with skin cancer. Cytotoxic T lymphocytes (CTLs) attack melanoma cells in an HLA-restricted and tumor antigen-specific manner. Several melanoma-associated tumor antigens have been identified. These antigens are suitable candidates for a vaccination therapy of melanoma. Dendritic cells (DCs) are antigen-presenting cells (APCs) specialized for the induction of a primary T-cell response. Mouse studies have demonstrated the potent capacity of DCs to induce antitumor immunity. In the present clinical pilot study, DCs were generated in the presence of granulocyte/macrophage-colony stimulating factor (GM-CSF) and interleukin 4 (IL-4) and were pulsed with tumor lysate or a cocktail of peptides known to be recognized by CTLs, depending on the patient's HLA haplotype. Keyhole limpet hemocyanin (KLH) was added as a CD4 helper antigen and immunological tracer molecule. Sixteen patients with advanced melanoma were immunized on an outpatient basis. Vaccination was well tolerated. No physical sign of autoimmunity was detected in any of the patients. DC vaccination induced delayed-type hypersensitivity (DTH) reactivity toward KLH in all patients, as well as a positive DTH reaction to peptide-pulsed DCs in 11 patients. Recruitment of peptide-specific CTLs to the DTH challenge site was also demonstrated. Therefore, antigen-specific immunity was induced during DC vaccination. Objective responses were evident in 5 out of 16 evaluated patients (two complete responses, three partial responses) with regression of metastases in various organs (skin, soft tissue, lung, pancreas) and one additional minor response. These data indicate that vaccination with autologous DCs generated from peripheral blood is a safe and promising approach in the treatment of metastatic melanoma. Further studies are necessary to demonstrate clinical effectiveness and impact on the survival of melanoma patients.
Collapse
Affiliation(s)
- F O Nestle
- Department of Dermatology, University of Zurich Medical School, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
140
|
Weller M, Bornemann A, Ständer M, Schabet M, Dichgans J, Meyermann R. Humoral immune response to p53 in malignant glioma. J Neurol 1998; 245:169-72. [PMID: 9553848 DOI: 10.1007/s004150050199] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
p53 immunoreactivity and humoral immune response to p53 were examined in 14 patients with malignant glioma, including 4 patients with leptomeningeal glioma cell dissemination. Twelve patients expressed p53 protein within the tumour tissue. p53 antibodies were detected in the serum in 2 of 14 patients but never in the cerebrospinal fluid (CSF). Soluble p53 protein was detected neither in serum nor in CSF of the glioma patients. CSF levels of the immunosuppressive cytokine, transforming growth factor (TGF)-beta, were elevated in the glioma patients, including those with a humoral response to p53. These preliminary findings raise the possibility of systemic humoral immune responses to antigens, including mutant p53, expressed by glioma cells in the central nervous system.
Collapse
Affiliation(s)
- M Weller
- Neurologische Klinik, Universität Tübingen, Germany.
| | | | | | | | | | | |
Collapse
|
141
|
Abstract
Dendritic cells (DC) are the most potent antigen-presenting cells. Thus, ex vivo antigen-pulsed DC are a potentially powerful tool to induce in vivo immunity against tumor-associated or viral antigens. Therefore, culture methods to generate high numbers of DC from bone marrow or blood CD34+ hematopoietic progenitor cells have recently been developed. These methods, which use different combinations of growth factor--mainly granulocyte/macrophage colony-stimulating factor (GM-CSF), tumor necrosis factor (TNF)-alpha and interleukin (IL)-4--make the characterization of DC obtained from CD34+ cells of different origins easier and allow to assess whether DC relate to a unique or distinct differentiation pathways. Monocytes and even macrophages can also directly differentiate into DC in the presence of GM-CSF and IL-4. This has to be reconciled with evidence supporting earlier branching off of the macrophage and DC lineages, and raises questions as to the identity of the latter lineage. Apart from DC of myeloid origin, DC may also originate from lymphoid progenitors. Because the capacity of DC to capture, process and present antigens is known to vary according to their differentiation stage, and lymphoid DC might behave differently from lymphoid DC in this respect, the definition of which type of DC to use for immunotherapy must be more precise, in order to avoid detrimental side effects or results. From a practical point of view, it is also necessary to define the most appropriate cytokine combinations and schedules thereof to optimize proliferation and differentiation of DC from different origins. These conditions should then be applied to generated DC for their efficient and safe use for clinical immunotherapy.
Collapse
Affiliation(s)
- J C Gluckman
- Service d'immunologie biologique, unité de biologie et thérapeutique des pathologies immunitaires et Laboratoire d'immunologie cellulaire de l'Ecole pratique des hautes études, hôpital de la Pitié-Salpêtrière, Paris, France
| | | | | |
Collapse
|
142
|
Anton D, Dabadghao S, Palucka K, Holm G, Yi Q. Generation of dendritic cells from peripheral blood adherent cells in medium with human serum. Scand J Immunol 1998; 47:116-21. [PMID: 9496685 DOI: 10.1046/j.1365-3083.1998.00284.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Dendritic cells (DC) provide an effective pathway for presenting antigens to T cells, both self-antigens during T-cell development and foreign antigens during immunity. As such, these cells may be promising adjuvants for immunotherapy. Thus, it is important to establish simple and fast method(s) to generate sufficient numbers of human DC in medium free of calf serum so that the cells can be used for both experimental and clinical purposes. In this report, we used peripheral blood adherent cells, without laborious cell purification or depletion, as the starting population and cultured them in medium supplemented with granulocyte/macrophage colony-stimulating factor and interleukin-4. Substantial numbers of cells with the phenotypical and functional characteristics of immature DC were obtained in a 7-day culture. We then compared DC cultured in medium supplemented with either fetal calf serum or pooled human ABRh+ serum and found no difference in cell yields and in their ability to stimulate alloreactive T cells or to present soluble antigens to T cells. Irradiated cells were less efficient than non-irradiated cells in antigen presentation and stimulation of T cells. Finally, we have examined DC with or without additional tumour necrosis factor-alpha treatment and found that antigen-pulsed mature cells could as efficiently present antigen to T cells as did immature cells. This method is suitable for the generation of DC in studies of large clinical materials.
Collapse
Affiliation(s)
- D Anton
- Center for Molecular Medicine, Department of Medicine, Karolinska Hospital, Stockholm, Sweden
| | | | | | | | | |
Collapse
|
143
|
Abstract
T-cells play an important role in in vivo tumor rejection in many animal tumor models and in human melanoma. Many human tumor antigens recognized by autologous T-cells have now been identified. These are found to be nonmutated and mutated peptides derived from various self proteins as well as viral proteins. A variety of mechanisms involved in generating these T-cell epitopes on growing cancers have also been identified. However, the role of these identified antigens remains to be evaluated. Passive or active immunotherapies using these identified tumor antigens are being conducted in many institutions. The results obtained from these clinical trials may give us better insight into the role of T-cell responses to each antigen in tumor rejection as well as the development of new antigen-specific immunotherapies for patients with cancer.
Collapse
Affiliation(s)
- Y Kawakami
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-1502, USA.
| | | |
Collapse
|
144
|
Morgan DJ, Kreuwel HTC, Fleck S, Levitsky HI, Pardoll DM, Sherman LA. Activation of Low Avidity CTL Specific for a Self Epitope Results in Tumor Rejection But Not Autoimmunity. THE JOURNAL OF IMMUNOLOGY 1998. [DOI: 10.4049/jimmunol.160.2.643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
To determine how self-tolerance can alter the ability of the immune system to respond against tumor-associated Ags that are also expressed by normal tissue, we designed experiments in which the same protein was expressed both as a tumor Ag and as a transgene product. Unlike conventional BALB/c mice that rejected renal carcinoma cells transfected with the influenza virus hemagglutinin (Renca-HA), transgenic mice that are tolerant of HA due to its expression as a self-Ag on pancreatic islet β cells, (Ins-HA mice) supported progressive growth of these tumor cells. However, when Ins-HA mice were immunized with a recombinant strain of vaccinia virus expressing the dominant H-2Kd peptide epitope of HA before receiving Renca-HA cells, they too were able to reject the tumor cells. Rejection of Renca-HA cells by immunized Ins-HA mice was found to be associated with the generation of CTL having much lower avidity for target cells presenting the KdHA epitope than CTL from immunized conventional BALB/c mice. Significantly, we show that self-tolerance to the HA Ag is quantitative rather then absolute, and that vaccination of Ins-HA mice can activate low avidity KdHA-specific CD8+ T cells that are able to reject tumor cells expressing high levels of HA, yet these mice remain tolerant of pancreatic islet β cells expressing HA.
Collapse
Affiliation(s)
- David J. Morgan
- *Department of Immunology, The Scripps Research Institute, La Jolla, CA 92037; and
| | - Huub T. C. Kreuwel
- *Department of Immunology, The Scripps Research Institute, La Jolla, CA 92037; and
| | - Shonna Fleck
- *Department of Immunology, The Scripps Research Institute, La Jolla, CA 92037; and
| | - Hyam I. Levitsky
- †Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Drew M. Pardoll
- †Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Linda A. Sherman
- *Department of Immunology, The Scripps Research Institute, La Jolla, CA 92037; and
| |
Collapse
|
145
|
Fujita H, Senju S, Yokomizo H, Saya H, Ogawa M, Matsushita S, Nishimura Y. Evidence that HLA class II-restricted human CD4+ T cells specific to p53 self peptides respond to p53 proteins of both wild and mutant forms. Eur J Immunol 1998; 28:305-16. [PMID: 9485210 DOI: 10.1002/(sici)1521-4141(199801)28:01<305::aid-immu305>3.0.co;2-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
By stimulating peripheral blood mononuclear cells of four healthy donors with a mixture of overlapping peptides representing the core domain of p53, we established two CD4+ alphabeta T cell clones and four lines that recognized wild-type and mutant p53 proteins as well as p53 self peptides in an HLA class II-restricted fashion. Two T cell lines established from two unrelated donors reacted to the p53 peptide (p)153-166 and p108-122, respectively, in the context of DP5 molecules. Two T cell clones established from two other unrelated donors were specific for p193-204 in the context of DRB1*1401 and for p153-165 in the context of DP5, respectively. These two T cell clones responded almost equally to both wild-type and four mutant recombinant p53 proteins. The proliferative responses of these T cell clones to p53 recombinant proteins were augmented by heat denaturing, thereby suggesting that altered conformation of the protein facilitates proteolytic processing to produce antigenic peptides. The DRB1*1401-restricted T cell clone specific for p193-204 killed a B lymphoblastoid cell line homozygous for HLA-DRB1*1401 when the cell line was pre-pulsed with p53 protein as well as peptide. These results indicate that CD4+ T cells reactive to p53 do exist in healthy individuals and the epitopes are probably ignored by the immune system under physiological conditions. It is suggested that such epitopes stimulate T cells to induce anti-p53 antibody production in cancer patients as previously reported by others. The possible involvement of p53-reactive T cells in anti-tumor immunity is discussed.
Collapse
Affiliation(s)
- H Fujita
- Department of Neuroscience and Immunology, Kumamoto University Graduate School of Medical Sciences, Japan
| | | | | | | | | | | | | |
Collapse
|
146
|
Hurpin C, Rotarioa C, Bisceglia H, Chevalier M, Tartaglia J, Erdile L. The mode of presentation and route of administration are critical for the induction of immune responses to p53 and antitumor immunity. Vaccine 1998; 16:208-15. [PMID: 9607032 DOI: 10.1016/s0264-410x(97)00190-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
We have examined the immune response to full-length wild-type human p53 presented by a recombinant canarypox vector (ALVAC) and by plasmid DNA. For the ALVAC recombinant, intravenous, but not subcutaneous, intramuscular or intradermal administration, induced CD8+ CTLs that lysed tumor cells transfected with human mutant p53. Intrasplenic administration also induced CTLs. Biodistribution studies showed that intravenously injected ALVAC localized primarily in the lung, liver and spleen, whereas intramuscularly injected virus remained predominantly at the injection site. Intradermal and intramuscular immunization with naked plasmid DNA encoding human wild-type p53 also induced a specific CTL response. DNA immunization induced complete protection against challenge with a mouse embryo fibroblast transfected with human mutant p53 and partial, but significant, protection against a transfected mastocytoma. The ALVAC recombinant induced partial protection in both models. These results suggest that recombinant ALVAC and DNA might be interesting presentation platforms for p53 to be tested in clinical studies.
Collapse
Affiliation(s)
- C Hurpin
- Pasteur Mérieux Connaught, Marcy L'Etoile, France
| | | | | | | | | | | |
Collapse
|
147
|
Gnjatic S, Cai Z, Viguier M, Chouaib S, Guillet JG, Choppin J. Accumulation of the p53 Protein Allows Recognition by Human CTL of a Wild-Type p53 Epitope Presented by Breast Carcinomas and Melanomas. THE JOURNAL OF IMMUNOLOGY 1998. [DOI: 10.4049/jimmunol.160.1.328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Abstract
The p53 protein is accumulated in tumor cells of many human cancers and can elicit in vivo humoral and proliferative responses. Rare reports about p53-mediated tumor recognition by CTLs have remained questioned. We therefore studied a panel of breast tumor and melanoma cell lines that we assayed for the presence of accumulated p53 and surface HLA-A2 and for the presentation of p53 epitopes. From PBMC of a healthy donor, we have generated a CTL line, D5/L9V, directed against HLA-A2-restricted peptide 264–272 from wild-type p53. It efficiently lysed breast adenocarcinomas MCF-7, MCF7/RA1, and MDA-MB-231, and melanoma M8, which all accumulate the p53 protein. Using competition assays, we made sure that tumor lysis by D5/L9V was due to recognition of endogenously produced p53 peptide 264–272 associated with the HLA-A2.1 molecule on the surface of these tumor cells. Cells with undetectable levels of wild-type p53, such as lymphoblastoid cells and melanoma M74, were not recognized by D5/L9V. Neither were breast tumor cell line MCF7/ADR nor melanoma line M44 because of HLA loss. This study therefore shows that it is possible to obtain in vitro CTL lines that specifically recognize a p53 epitope spontaneously presented by a variety of HLA-A2+ transformed cell lines provided they display abnormal patterns of p53 expression. This work points out that breast tumors and melanomas share a p53 epitope, and raises hopes for future immunotherapeutic approaches.
Collapse
Affiliation(s)
- Sacha Gnjatic
- *INSERM U445, Institut Cochin de Génétique Moléculaire, Université René Descartes Paris V, Paris, France; and
| | - Zhenzi Cai
- †INSERM CJF 94-11, Institut Gustave Roussy, Ville-Juif, France
| | - Mireille Viguier
- *INSERM U445, Institut Cochin de Génétique Moléculaire, Université René Descartes Paris V, Paris, France; and
| | - Salem Chouaib
- †INSERM CJF 94-11, Institut Gustave Roussy, Ville-Juif, France
| | - Jean-Gérard Guillet
- *INSERM U445, Institut Cochin de Génétique Moléculaire, Université René Descartes Paris V, Paris, France; and
| | - Jeannine Choppin
- *INSERM U445, Institut Cochin de Génétique Moléculaire, Université René Descartes Paris V, Paris, France; and
| |
Collapse
|
148
|
McCarty TM, Yu Z, Liu X, Diamond DJ, Ellenhorn JD. An HLA-restricted, p53 specific immune response from HLA transgenic p53 knockout mice. Ann Surg Oncol 1998; 5:93-9. [PMID: 9524714 DOI: 10.1007/bf02303770] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND p53 is over-expressed in most human malignancies and is therefore an attractive target for immunotherapy. Unfortunately, a human cytotoxic T cell response to p53 is difficult to generate. p53 knockout transgenic mice may provide a model to circumvent immunologic tolerance to p53 and develop high-affinity p53-specific cytotoxic T lymphocytes (CTL). METHODS p53 knockout, HLA A2.1 transgenic mice were generated and immunized with the immunodominant wild-type p53 nonamer peptide epitope p53149-157. Two weeks later splenocytes were harvested and stimulated in vitro with acid-treated, p53 peptide-pulsed syngeneic blast cells. Cultures were restimulated weekly with acid-treated, p53 peptide-pulsed Jurkat cells transfected with the HLA A2.1 gene. Peptide-specific cytotoxic activity was measured by chromium release assay, and the resulting CD8+ effectors were cloned via limiting dilution. RESULTS P53 peptide-specific CTL were generated against p53149-157. Clones generated from the p53149-157 cell line demonstrated high affinity and specificity for p53149-157 when presented by HLA A2.1+ antigen-presenting cells. The p53149-157 CTL killed only cells overexpressing p53 cells that were HLA A2.1+ and did not kill cells with normal levels of p53 expression or those that were HLA A2.1-. CONCLUSION HLA transgenic mice not previously exposed to the p53 protein provide a useful model for generating high-affinity p53-specific CTL.
Collapse
Affiliation(s)
- T M McCarty
- Department of Surgery, City of Hope National Medical Center, Duarte, CA 91010, USA
| | | | | | | | | |
Collapse
|
149
|
|
150
|
Specht JM, Wang G, Do MT, Lam JS, Royal RE, Reeves ME, Rosenberg SA, Hwu P. Dendritic cells retrovirally transduced with a model antigen gene are therapeutically effective against established pulmonary metastases. J Exp Med 1997; 186:1213-21. [PMID: 9334360 PMCID: PMC2199086 DOI: 10.1084/jem.186.8.1213] [Citation(s) in RCA: 246] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Dendritic cells (DCs) are bone marrow-derived leukocytes that function as potent antigen presenting cells capable of initiating T cell-dependent responses from quiescent lymphocytes. DC pulsed with tumor-associated antigen (TAA) peptide or protein have recently been demonstrated to elicit antigen-specific protective antitumor immunity in a number of murine models. Transduction of DCs with TAA genes may allow stable, prolonged antigen expression as well as the potential for presentation of multiple, or unidentified, epitopes in association with major histocompatibility complex class I and/or class II molecules. To evaluate the potential efficacy of retrovirally transduced DCs, bone marrow cells harvested from BALB/c mice were transduced with either a model antigen gene encoding beta-galactosidase (beta-gal) or a control gene encoding rat HER-2/neu (Neu) by coculture with irradiated ecotropic retroviral producer lines. Bone marrow cells were differentiated into DC in vitro using granulocyte/macrophage colony-stimulating factor and interleukin-4. After 7 d in culture, cells were 45-78% double positive for DC phenotypic cell surface markers by FACS(R) analysis, and DC transduced with beta-gal were 41-72% positive for beta-gal expression by X-gal staining. In addition, coculture of beta-gal transduced DC with a beta-gal-specific T cell line (CTLx) resulted in the production of large amounts of interferon-gamma, demonstrating that transduced DCs could process and present endogenously expressed beta-gal. DC transduced with beta-gal and control rat HER-2/neu were then used to treat 3-d lung metastases in mice bearing an experimental murine tumor CT26.CL25, expressing the model antigen, beta-gal. Treatment with beta-gal-transduced DC significantly reduced the number of pulmonary metastatic nodules compared with treatment with Hank's balanced salt solution or DCs transduced with rat HER-2/neu. In addition, immunization with beta-gal-transduced DCs resulted in the generation of antigen-specific cytotoxic T lymphocytes (CTLs), which were significantly more reactive against relevant tumor targets than CTLs generated from mice immunized with DCs pulsed with the Ld-restricted beta-gal peptide. The results observed in this rapidly lethal tumor model suggest that DCs transduced with TAA may be a useful treatment modality in tumor immunotherapy.
Collapse
Affiliation(s)
- J M Specht
- Surgery Branch, National Cancer Institute, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | | | |
Collapse
|