1
|
Alarcon NO, Jaramillo M, Mansour HM, Sun B. Therapeutic Cancer Vaccines—Antigen Discovery and Adjuvant Delivery Platforms. Pharmaceutics 2022; 14:pharmaceutics14071448. [PMID: 35890342 PMCID: PMC9325128 DOI: 10.3390/pharmaceutics14071448] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/28/2022] [Accepted: 06/30/2022] [Indexed: 12/15/2022] Open
Abstract
For decades, vaccines have played a significant role in protecting public and personal health against infectious diseases and proved their great potential in battling cancers as well. This review focused on the current progress of therapeutic subunit vaccines for cancer immunotherapy. Antigens and adjuvants are key components of vaccine formulations. We summarized several classes of tumor antigens and bioinformatic approaches of identification of tumor neoantigens. Pattern recognition receptor (PRR)-targeting adjuvants and their targeted delivery platforms have been extensively discussed. In addition, we emphasized the interplay between multiple adjuvants and their combined delivery for cancer immunotherapy.
Collapse
Affiliation(s)
- Neftali Ortega Alarcon
- Skaggs Pharmaceutical Sciences Center, College of Pharmacy, The University of Arizona, Tucson, AZ 85721, USA; (N.O.A.); (M.J.); (H.M.M.)
| | - Maddy Jaramillo
- Skaggs Pharmaceutical Sciences Center, College of Pharmacy, The University of Arizona, Tucson, AZ 85721, USA; (N.O.A.); (M.J.); (H.M.M.)
| | - Heidi M. Mansour
- Skaggs Pharmaceutical Sciences Center, College of Pharmacy, The University of Arizona, Tucson, AZ 85721, USA; (N.O.A.); (M.J.); (H.M.M.)
- The University of Arizona Cancer Center, Tucson, AZ 85721, USA
- Department of Medicine, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
- BIO5 Institute, The University of Arizona, Tucson, AZ 85721, USA
| | - Bo Sun
- Skaggs Pharmaceutical Sciences Center, College of Pharmacy, The University of Arizona, Tucson, AZ 85721, USA; (N.O.A.); (M.J.); (H.M.M.)
- The University of Arizona Cancer Center, Tucson, AZ 85721, USA
- BIO5 Institute, The University of Arizona, Tucson, AZ 85721, USA
- Correspondence: ; Tel.: +1-520-621-6420
| |
Collapse
|
2
|
Agupitan AD, Neeson P, Williams S, Howitt J, Haupt S, Haupt Y. P53: A Guardian of Immunity Becomes Its Saboteur through Mutation. Int J Mol Sci 2020; 21:E3452. [PMID: 32414156 PMCID: PMC7278985 DOI: 10.3390/ijms21103452] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 05/06/2020] [Accepted: 05/11/2020] [Indexed: 02/06/2023] Open
Abstract
Awareness of the importance of immunity in controlling cancer development triggered research into the impact of its key oncogenic drivers on the immune response, as well as their value as targets for immunotherapy. At the heart of tumour suppression is p53, which was discovered in the context of viral infection and now emerges as a significant player in normal and cancer immunity. Wild-type p53 (wt p53) plays fundamental roles in cancer immunity and inflammation. Mutations in p53 not only cripple wt p53 immune functions but also sinisterly subvert the immune function through its neomorphic gain-of-functions (GOFs). The prevalence of mutant p53 across different types of human cancers, which are associated with inflammatory and immune dysfunction, further implicates mutant p53 in modulating cancer immunity, thereby promoting tumorigenesis, metastasis and invasion. In this review, we discuss several mutant p53 immune GOFs in the context of the established roles of wt p53 in regulating and responding to tumour-associated inflammation, and regulating innate and adaptive immunity. We discuss the capacity of mutant p53 to alter the tumour milieu to support immune dysfunction, modulate toll-like receptor (TLR) signalling pathways to disrupt innate immunity and subvert cell-mediated immunity in favour of immune privilege and survival. Furthermore, we expose the potential and challenges associated with mutant p53 as a cancer immunotherapy target and underscore existing therapies that may benefit from inquiry into cancer p53 status.
Collapse
Affiliation(s)
- Arjelle Decasa Agupitan
- Tumour Suppression Laboratory, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne 3000, Victoria, Australia; (A.D.A.); (S.H.)
| | - Paul Neeson
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville 3010, Victoria, Australia;
- Cancer Immunology Research, Peter MacCallum Cancer Centre, Melbourne 3000, Victoria, Australia
| | - Scott Williams
- Division of Radiation Oncology and Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne 3000, Victoria, Australia;
| | - Jason Howitt
- School of Health Sciences, Swinburne University, Melbourne 3122, Victoria, Australia;
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville 3010, Victoria, Australia
| | - Sue Haupt
- Tumour Suppression Laboratory, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne 3000, Victoria, Australia; (A.D.A.); (S.H.)
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville 3010, Victoria, Australia;
| | - Ygal Haupt
- Tumour Suppression Laboratory, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne 3000, Victoria, Australia; (A.D.A.); (S.H.)
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville 3010, Victoria, Australia;
- Department of Clinical Pathology, University of Melbourne, Parkville 3010, Victoria, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne 3800, Victoria, Australia
| |
Collapse
|
3
|
Lee JM, Lee MH, Garon E, Goldman JW, Salehi-Rad R, Baratelli FE, Schaue D, Wang G, Rosen F, Yanagawa J, Walser TC, Lin Y, Park SJ, Adams S, Marincola FM, Tumeh PC, Abtin F, Suh R, Reckamp KL, Lee G, Wallace WD, Lee S, Zeng G, Elashoff DA, Sharma S, Dubinett SM. Phase I Trial of Intratumoral Injection of CCL21 Gene-Modified Dendritic Cells in Lung Cancer Elicits Tumor-Specific Immune Responses and CD8 + T-cell Infiltration. Clin Cancer Res 2017; 23:4556-4568. [PMID: 28468947 DOI: 10.1158/1078-0432.ccr-16-2821] [Citation(s) in RCA: 143] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 03/27/2017] [Accepted: 04/26/2017] [Indexed: 01/15/2023]
Abstract
Purpose: A phase I study was conducted to determine safety, clinical efficacy, and antitumor immune responses in patients with advanced non-small cell lung carcinoma (NSCLC) following intratumoral administration of autologous dendritic cells (DC) transduced with an adenoviral (Ad) vector expressing the CCL21 gene (Ad-CCL21-DC). We evaluated safety and tumor antigen-specific immune responses following in situ vaccination (ClinicalTrials.gov: NCT01574222).Experimental Design: Sixteen stage IIIB/IV NSCLC subjects received two vaccinations (1 × 106, 5 × 106, 1 × 107, or 3 × 107 DCs/injection) by CT- or bronchoscopic-guided intratumoral injections (days 0 and 7). Immune responses were assessed by tumor antigen-specific peripheral blood lymphocyte induction of IFNγ in ELISPOT assays. Tumor biopsies were evaluated for CD8+ T cells by IHC and for PD-L1 expression by IHC and real-time PCR (RT-PCR).Results: Twenty-five percent (4/16) of patients had stable disease at day 56. Median survival was 3.9 months. ELISPOT assays revealed 6 of 16 patients had systemic responses against tumor-associated antigens (TAA). Tumor CD8+ T-cell infiltration was induced in 54% of subjects (7/13; 3.4-fold average increase in the number of CD8+ T cells per mm2). Patients with increased CD8+ T cells following vaccination showed significantly increased PD-L1 mRNA expression.Conclusions: Intratumoral vaccination with Ad-CCL21-DC resulted in (i) induction of systemic tumor antigen-specific immune responses; (ii) enhanced tumor CD8+ T-cell infiltration; and (iii) increased tumor PD-L1 expression. Future studies will evaluate the role of combination therapies with PD-1/PD-L1 checkpoint inhibition combined with DC-CCL21 in situ vaccination. Clin Cancer Res; 23(16); 4556-68. ©2017 AACR.
Collapse
Affiliation(s)
- Jay M Lee
- Lung Cancer Research Program, Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Los Angeles, California. .,Department of Surgery, Division of Thoracic Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Mi-Heon Lee
- Lung Cancer Research Program, Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Los Angeles, California.,Department of Surgery, Division of Thoracic Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Edward Garon
- Department of Medicine, Division of Hematology and Oncology, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Jonathan W Goldman
- Department of Medicine, Division of Hematology and Oncology, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Ramin Salehi-Rad
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Felicita E Baratelli
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Dörthe Schaue
- Department of Radiology, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Gerald Wang
- Lung Cancer Research Program, Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Los Angeles, California.,Department of Medicine, Division of Pulmonary and Critical Care Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Fran Rosen
- Lung Cancer Research Program, Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Los Angeles, California.,Department of Medicine, Division of Pulmonary and Critical Care Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Jane Yanagawa
- Lung Cancer Research Program, Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Los Angeles, California.,Department of Surgery, Division of Thoracic Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Tonya C Walser
- Lung Cancer Research Program, Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Los Angeles, California.,Department of Medicine, Division of Pulmonary and Critical Care Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Ying Lin
- Lung Cancer Research Program, Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Los Angeles, California.,Department of Medicine, Division of Pulmonary and Critical Care Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Stacy J Park
- Lung Cancer Research Program, Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Los Angeles, California.,Department of Medicine, Division of Pulmonary and Critical Care Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Sharon Adams
- Department of Transfusion Medicine, NIH, Bethesda, Maryland
| | | | - Paul C Tumeh
- Lung Cancer Research Program, Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Los Angeles, California.,Department of Dermatology, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Fereidoun Abtin
- Department of Radiology, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Robert Suh
- Department of Radiology, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Karen L Reckamp
- Department of Medical Oncology and Therapeutics Research, City of Hope, Duarte, California
| | - Gina Lee
- Lung Cancer Research Program, Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Los Angeles, California.,Department of Medicine, Division of Pulmonary and Critical Care Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California.,Department of Medicine, Division of Pulmonary and Critical Care Medicine, Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California
| | - William D Wallace
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Sarah Lee
- Department of Urology, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Gang Zeng
- Department of Urology, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - David A Elashoff
- Lung Cancer Research Program, Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Los Angeles, California.,Department of Biostatistics, Division of General Internal Medicine and Health Services Research, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Sherven Sharma
- Lung Cancer Research Program, Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Los Angeles, California.,Department of Medicine, Division of Pulmonary and Critical Care Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California.,Molecular Gene Medicine Laboratory, Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California
| | - Steven M Dubinett
- Lung Cancer Research Program, Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Los Angeles, California. .,Department of Medicine, Division of Pulmonary and Critical Care Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California.,Department of Medical Oncology and Therapeutics Research, City of Hope, Duarte, California.,Department of Medicine, Division of Pulmonary and Critical Care Medicine, Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California.,Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California
| |
Collapse
|
4
|
Protti MP, De Monte L, Monte LD, Di Lullo G, Lullo GD. Tumor antigen-specific CD4+ T cells in cancer immunity: from antigen identification to tumor prognosis and development of therapeutic strategies. ACTA ACUST UNITED AC 2014; 83:237-46. [PMID: 24641502 DOI: 10.1111/tan.12329] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Indexed: 12/22/2022]
Abstract
CD4(+) T cells comprise a large fraction of tumor infiltrating lymphocytes and it is now established that they may exert an important role in tumor immune-surveillance. Several CD4(+) T cell subsets [i.e. T helper (Th)1, Th2, T regulatory (Treg), Th17, Th22 and follicular T helper (Tfh)] have been described and differentiation of each subset depends on both the antigen presenting cells responsible for its activation and the cytokine environment present at the site of priming. Tumor antigen-specific CD4(+) T cells with different functional activity have been found in the blood of cancer patients and different CD4(+) T cell subsets have been identified at the tumor site by the expression of specific transcription factors and the profile of secreted cytokines. Importantly, depending on the subset, CD4(+) T cells may exert antitumor versus pro-tumor functions. Here we review the studies that first identified the presence of tumor-specific CD4(+) T cells in cancer patients, the techniques used to identify the tumor antigens recognized, the role of the different CD4(+) T cell subsets in tumor immunity and in cancer prognosis and the development of therapeutic strategies aimed at activating efficient antitumor CD4(+) T cell effectors.
Collapse
Affiliation(s)
- M P Protti
- Tumor Immunology Unit, San Raffaele Scientific Institute, Milan, Italy; Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| | | | | | | | | |
Collapse
|
5
|
Kumai T, Ishibashi K, Oikawa K, Matsuda Y, Aoki N, Kimura S, Hayashi S, Kitada M, Harabuchi Y, Celis E, Kobayashi H. Induction of tumor-reactive T helper responses by a posttranslational modified epitope from tumor protein p53. Cancer Immunol Immunother 2014; 63:469-78. [PMID: 24633296 PMCID: PMC11028558 DOI: 10.1007/s00262-014-1533-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Accepted: 03/03/2014] [Indexed: 12/22/2022]
Abstract
Posttranslational modifications regulate the function and stability of proteins, and the immune system is able to recognize some of these modifications. Therefore, the presence of posttranslational modifications increases the diversity of potential immune responses to a determinant antigen. The stimulation of tumor-specific CD4(+) helper T lymphocytes (HTLs) is considered important for the production of anti-tumor antibodies by B cells and for the generation and persistence of CD8(+) cytotoxic T lymphocytes, and in some instances, HTLs can directly reduce tumor cell growth. Identification of MHC class II-restricted peptide epitopes from tumor-associated antigens including those generated from posttranslational protein modifications should enable the improvement of peptide-based cancer immunotherapy. We describe here an MHC class II binding peptide from the tumor protein p53, which possesses an acetylated lysine at position 120 (p53110-124/AcK120) that is effective in eliciting CD4(+) T cell responses specific for the acetylated peptide. Most importantly, the acetylated peptide-reactive CD4 HTLs recognized the corresponding naturally processed posttranslational modified epitope presented by either dendritic cells loaded with tumor cell lysates or directly on tumors expressing p53 and the restricting MHC class II molecules. Treatment of tumor cells with a histone deacetylase inhibitor augmented their recognition by the p53110-124/AcK120-reactive CD4(+) T cells. These findings prove that the epitope p53110-124/AcK120 is immunogenic for anti-tumor responses and is likely to be useful for cancer immunotherapy.
Collapse
Affiliation(s)
- Takumi Kumai
- Department of Pathology, Asahikawa Medical University, Midorigaoka-Higashi 2-1-1, Asahikawa, 078-8510 Japan
- Department of Otolaryngology, Head and Neck Surgery, Asahikawa Medical University, Asahikawa, 078-8510 Japan
| | - Kei Ishibashi
- Department of Pathology, Asahikawa Medical University, Midorigaoka-Higashi 2-1-1, Asahikawa, 078-8510 Japan
- Respiratory and Breast Center, Asahikawa Medical University Hospital, Asahikawa, 078-8510 Japan
| | - Kensuke Oikawa
- Department of Pathology, Asahikawa Medical University, Midorigaoka-Higashi 2-1-1, Asahikawa, 078-8510 Japan
| | - Yoshinari Matsuda
- Respiratory and Breast Center, Asahikawa Medical University Hospital, Asahikawa, 078-8510 Japan
| | - Naoko Aoki
- Department of Pathology, Asahikawa Medical University, Midorigaoka-Higashi 2-1-1, Asahikawa, 078-8510 Japan
| | - Shoji Kimura
- Department of Pathology, Asahikawa Medical University, Midorigaoka-Higashi 2-1-1, Asahikawa, 078-8510 Japan
| | - Satoshi Hayashi
- Respiratory and Breast Center, Asahikawa Medical University Hospital, Asahikawa, 078-8510 Japan
| | - Masahiro Kitada
- Respiratory and Breast Center, Asahikawa Medical University Hospital, Asahikawa, 078-8510 Japan
| | - Yasuaki Harabuchi
- Department of Otolaryngology, Head and Neck Surgery, Asahikawa Medical University, Asahikawa, 078-8510 Japan
| | - Esteban Celis
- Cancer Immunology, Inflammation and Tolerance Program, Department of Medicine, Georgia Regents University Cancer Center, 1410 Laney Walker Boulevard, CN-4121, Augusta, GA 30912 USA
| | - Hiroya Kobayashi
- Department of Pathology, Asahikawa Medical University, Midorigaoka-Higashi 2-1-1, Asahikawa, 078-8510 Japan
| |
Collapse
|
6
|
Karyampudi L, Krco CJ, Kalli KR, Erskine CL, Hartmann LC, Goodman K, Ingle JN, Maurer MJ, Nassar A, Yu C, Disis ML, Wettstein PJ, Fikes JD, Beebe M, Ishioka G, Knutson KL. Identification of a broad coverage HLA-DR degenerate epitope pool derived from carcinoembryonic antigen. Cancer Immunol Immunother 2009; 59:161-71. [PMID: 19621224 DOI: 10.1007/s00262-009-0738-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2009] [Accepted: 07/02/2009] [Indexed: 12/19/2022]
Abstract
CD4 T cells are important for anti-tumor immune responses. Aside from their role in the activation of CD8 T cells, CD4 T cells also mediate anti-tumor immune responses by recruiting innate immune effectors into the tumor microenvironment. Thus, the search for strategies to boost CD4 T cell immunity is an active area of research. Our goal in this study was to identify HLA-DR epitopes of carcinoembryonic antigen (CEA), a commonly over-expressed tumor antigen. HLA-DR epitopes of CEA were identified using the epitope prediction program, PIC (predicted IC(50)) and tested using in vitro HLA-DR binding assays. Following CEA epitope confirmation, IFN-gamma ELIspot assays were used to detect existing immunity against the HLA-DR epitope panel of CEA in breast and ovarian cancer patients. In vitro generated peptide-specific CD4 T cells were used to determine whether the epitopes are naturally processed from CEA protein. Forty-three epitopes of CEA were predicted, 15 of which had high binding affinity for 8 or more common HLA-DR molecules. A degenerate pool of four, HLA-DR restricted 15 amino acid epitopes (CEA.24, CEA.176/354, CEA.488, and CEA.653) consisting of two novel epitopes (CEA.24 and CEA.488) was identified against which 40% of breast and ovarian cancer patients had pre-existent T cell immunity. All four epitopes are naturally processed by antigen-presenting cells. Hardy-Weinberg analysis showed that the pool is useful in approximately 94% of patients. Patients with breast or ovarian cancer demonstrate pre-existent immune responses to the tumor antigen CEA. The degenerate pool of CEA peptides may be useful for augmenting CD4 T cell immunity.
Collapse
Affiliation(s)
- Lavakumar Karyampudi
- Department of Immunology, College of Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Speetjens FM, Kuppen PJ, Welters MJ, Essahsah F, Voet van den Brink AME, Lantrua MGK, Valentijn ARP, Oostendorp J, Fathers LM, Nijman HW, Drijfhout JW, van de Velde CJ, Melief CJ, van der Burg SH. Induction of p53-Specific Immunity by a p53 Synthetic Long Peptide Vaccine in Patients Treated for Metastatic Colorectal Cancer. Clin Cancer Res 2009; 15:1086-95. [DOI: 10.1158/1078-0432.ccr-08-2227] [Citation(s) in RCA: 130] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
8
|
Chikamatsu K, Sakakura K, Takahashi G, Okamoto A, Furuya N, Whiteside TL, DeLeo AB, Masuyama K. CD4+ T cell responses to HLA-DP5-restricted wild-type sequence p53 peptides in patients with head and neck cancer. Cancer Immunol Immunother 2009; 58:1441-8. [PMID: 19184003 DOI: 10.1007/s00262-009-0661-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2008] [Accepted: 01/09/2009] [Indexed: 10/21/2022]
Abstract
Wild-type sequence (wt) p53 peptides are attractive candidates for broadly applicable cancer vaccines. Evidence has been accumulating which indicates that CD4+ Th cells have an important role in generating and maintaining antitumor immune responses. To elucidate the nature of CD4+ Th responses to wt p53 epitopes in patients with squamous cell carcinoma of the head and neck (SCCHN), peripheral blood mononuclear cells (PBMCs) from HLA-DP5+ patients were stimulated with HLA-DP5-restricted wt p53 peptides, p53(108-122) or p53(153-166), and tested for the release of IFN-gamma and IL-5 in ELISPOT assays. Immunohistochemistry for p53 accumulation in tumors, and ELISA for serum antibodies to p53 were also performed. Eleven (57.9%) of 19 HLA-DP5+ patients but none of 5 healthy donors had detectable Th1 and/or Th2 responses to wt p53 peptides by ELISPOT assay. Among these 11 responding patients, 9 (81.8%) and all 11 (100%) patients had a tumor burden and p53 accumulation, respectively. On the other hand, two responding patients were in post-operative condition. Interestingly, among nine patients with a tumor burden, four patients with early disease showed either Th1-polarized or mixed Th1/Th2 responses, while five patients with advanced disease showed either Th2-polarized or mixed Th1/Th2 responses. Our results suggest that wt p53(108-122) and p53(153-166) peptides stimulate both Th1- and Th2-type CD4+ T cell responses in patients with SCCHN, and anti-p53 Th responses may persist even after surgical resection of the tumor; however, the presence of a tumor and its progression may affect the nature of immune responses to wt p53 peptides.
Collapse
Affiliation(s)
- Kazuaki Chikamatsu
- Department of Otolaryngology-Head and Neck Surgery, Gunma University Graduate School of Medicine, Maebashi, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
9
|
DeLeo AB, Whiteside TL. Development of multi-epitope vaccines targeting wild-type sequence p53 peptides. Expert Rev Vaccines 2008; 7:1031-40. [PMID: 18767952 DOI: 10.1586/14760584.7.7.1031] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Loss of p53 tumor-suppressor function is the most common abnormality in human cancer, which can result in enhanced presentation to immune cells of wild-type (wt)-sequence peptides from tumor p53 molecules, thus providing the rationale for wt p53 peptide-based cancer vaccines. We review evidence from preclinical murine tumor models and preclinical studies that led to the clinical introduction of wt p53 peptide-based vaccines for cancer immunotherapy. Overall, this review illustrates the complex process of wt p53 epitope selection and the issues and concerns involved in the application of p53-based vaccines for patients with cancer.
Collapse
Affiliation(s)
- Albert B DeLeo
- Department of Pathology, University of Pittsburgh School of Medicine and University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | | |
Collapse
|
10
|
James EA, Moustakas AK, Berger D, Huston L, Papadopoulos GK, Kwok WW. Definition of the peptide binding motif within DRB1*1401 restricted epitopes by peptide competition and structural modeling. Mol Immunol 2008; 45:2651-9. [PMID: 18276010 DOI: 10.1016/j.molimm.2007.12.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2007] [Accepted: 12/20/2007] [Indexed: 11/26/2022]
Abstract
This study identified the peptide-binding motif of HLA-DRA/DRB1*1401 (DR1401). First, peptides containing DR1401 restricted epitopes were identified using tetramer-guided epitope mapping. Among these, an influenza B peptide was selected for the motif study. After confirming the binding register for this peptide using a set of arginine substitutions, binding affinities were determined for 33 peptides derived from this influenza B sequence with single amino acid substitutions. The DR1401 peptide-binding motif was deduced from the relative binding affinities of these peptides and confirmed by structural modeling. Pocket 1 demonstrated a preference for aliphatic anchor residues and methionine. Pocket 4 accommodated methionine and aliphatic residues, but also allowed some polar and charged amino acids. Pocket 6 preferred basic residues but also allowed some polar and aliphatic amino acids. Pocket 9 preferred aliphatic and aromatic amino acids and tolerated some polar residues but excluded all charged residues. Together these preferences define a distinct set of peptides that can be presented by DR1401. The resulting motif was used to verify T cell epitopes within the novel antigenic peptides identified by tetramer-guided epitope mapping and within peptides from published reports that contain putative DR1401 epitopes.
Collapse
Affiliation(s)
- Eddie A James
- Benaroya Research Institute at Virginia Mason, Seattle, WA 98101, USA
| | | | | | | | | | | |
Collapse
|
11
|
Senju S, Suemori H, Zembutsu H, Uemura Y, Hirata S, Fukuma D, Matsuyoshi H, Shimomura M, Haruta M, Fukushima S, Matsunaga Y, Katagiri T, Nakamura Y, Furuya M, Nakatsuji N, Nishimura Y. Genetically Manipulated Human Embryonic Stem Cell-Derived Dendritic Cells with Immune Regulatory Function. Stem Cells 2007; 25:2720-9. [PMID: 17690179 DOI: 10.1634/stemcells.2007-0321] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Genetically manipulated dendritic cells (DC) are considered to be a promising means for antigen-specific immune therapy. This study reports the generation, characterization, and genetic modification of DC derived from human embryonic stem (ES) cells. The human ES cell-derived DC (ES-DC) expressed surface molecules typically expressed by DC and had the capacities to stimulate allogeneic T lymphocytes and to process and present protein antigen in the context of histocompatibility leukocyte antigen (HLA) class II molecule. Genetic modification of human ES-DC can be accomplished without the use of viral vectors, by the introduction of expression vector plasmids into undifferentiated ES cells by electroporation and subsequent induction of differentiation of the transfectant ES cell clones to ES-DC. ES-DC introduced with invariant chain-based antigen-presenting vectors by this procedure stimulated HLA-DR-restricted antigen-specific T cells in the absence of exogenous antigen. Forced expression of programmed death-1-ligand-1 in ES-DC resulted in the reduction of the proliferative response of allogeneic T cells cocultured with the ES-DC. Generation and genetic modification of ES-DC from nonhuman primate (cynomolgus monkey) ES cells was also achieved by the currently established method. ES-DC technology is therefore considered to be a novel means for immune therapy.
Collapse
Affiliation(s)
- Satoru Senju
- Department of Immunogenetics, Graduate School of Medical and Pharmaceutical Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Lambeck A, Leffers N, Hoogeboom BN, Sluiter W, Hamming I, Klip H, ten Hoor K, Esajas M, van Oven M, Drijfhout JW, Platteel I, Offringa R, Hollema H, Melief K, van der Burg S, van der Zee A, Daemen T, Nijman H. P53-specific T cell responses in patients with malignant and benign ovarian tumors: implications for p53 based immunotherapy. Int J Cancer 2007; 121:606-14. [PMID: 17415711 DOI: 10.1002/ijc.22710] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Despite intensive treatment, 70% of the ovarian cancer patients will develop recurrent disease, emphasizing the need for new approaches such as immunotherapy. A promising antigenic target for immunotherapy in ovarian cancer is the frequently overexpressed p53 protein. The aim of the study was to evaluate the nature and magnitude of the baseline anti-p53 immune response in ovarian cancer patients. P53-specific T cell responses were detected in both half of the ovarian cancer patients as in the group of control subjects, consisting of women with benign ovarian tumors and healthy controls. Importantly, while in the control group p53-specific immunity was detected among the CD45RA(+) naïve subset of T cells only, the p53-specific T-cell responses in ovarian cancer patients were also present in the CD45RO(+) memory T-cell subset, suggesting that in the cancer patients sufficient amounts of cancer-derived p53 was presented to induce the formation of a p53-specific memory T-cell response. Further characterization of the p53-specific memory T-cell responses revealed that in addition to the type 1 cytokine IFN-gamma also the type 2 cytokines IL-4 and IL-5, as well as the immunosuppressive cytokine IL-10 were produced. Notably, p53-specific T cells were not only detected in the peripheral blood, but also among tumor infiltrating lymphocytes and in tumor-draining lymph nodes. In conclusion, the existence of a weak mixed T-helper type 1 and 2 p53-specific T-cell repertoire supports the rationale of using p53 long peptides in vaccination strategies aiming at the induction of p53-specific Th1/CTL immunity.
Collapse
Affiliation(s)
- Annechien Lambeck
- Department of Gynaecology, University Medical Center Groningen, University of Groningen, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Gouttefangeas C, Stenzl A, Stevanović S, Rammensee HG. Immunotherapy of renal cell carcinoma. Cancer Immunol Immunother 2007; 56:117-28. [PMID: 16676181 PMCID: PMC11030119 DOI: 10.1007/s00262-006-0172-4] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2006] [Accepted: 04/10/2006] [Indexed: 01/02/2023]
Abstract
Carcinomas of the kidney generally have a poor prognosis and respond minimally to classical radiotherapy or chemotherapy. Immunotherapy constitutes an interesting alternative to these established forms of treatment, and indeed, cytokine-based therapies have been used for many years, leading to favorable clinical responses in a small subset of patients. During the past few years, immunotherapeutical trials targeting renal cell tumor-associated antigens have also been reported, with diverse passive or active approaches using antibodies or aimed at activating tumor-directed T lymphocytes. The following review presents the results and the progress made in the field, including classical cytokine treatments, non-myeloablative stem cell transplantation and antigen specific-based trials, with special focus on T-cell studies. In consideration of the few specific molecular targets described so far for this tumor entity, current strategies which can lead to the identification of new relevant antigens will be discussed. Hopefully these will very soon contribute to an improvement in renal cell carcinoma specific immunotherapy and its evaluation.
Collapse
Affiliation(s)
- Cécile Gouttefangeas
- Institute for Cell Biology, Department of Immunology, Eberhard Karls University, Auf der Morgenstelle 15, 72076, Tubingen, Germany.
| | | | | | | |
Collapse
|
14
|
Yokomine K, Nakatsura T, Minohara M, Kira JI, Kubo T, Sasaki Y, Nishimura Y. Immunization with heat shock protein 105-pulsed dendritic cells leads to tumor rejection in mice. Biochem Biophys Res Commun 2006; 343:269-78. [PMID: 16540092 DOI: 10.1016/j.bbrc.2006.02.142] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2006] [Accepted: 02/21/2006] [Indexed: 11/18/2022]
Abstract
Recently, we reported that heat shock protein 105 (HSP105) DNA vaccination induced anti-tumor immunity. In this study, we set up a preclinical study to investigate the usefulness of dendritic cells (DCs) pulsed with mouse HSP105 as a whole protein for cancer immunotherapy in vivo. The recombinant HSP105 did not induce DC maturation, and the mice vaccinated with HSP105-pulsed BM-DCs were markedly prevented from the growth of subcutaneous tumors, accompanied with a massive infiltration of both CD4+ T cells and CD8+ T cells into the tumors. In depletion experiments, we proved that both CD4+ T cells and CD8+ T cells play a crucial role in anti-tumor immunity. Both CD4+ T cells and CD8+ T cells specific to HSP105 were induced by stimulation with HSP105-pulsed DCs. As a result, vaccination of mice with BM-DCs pulsed with HSP105 itself could elicit a stronger tumor rejection in comparison to DNA vaccination.
Collapse
Affiliation(s)
- Kazunori Yokomine
- Department of Immunogenetics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | | | | | | | | | | | | |
Collapse
|
15
|
Nijman HW, Lambeck A, van der Burg SH, van der Zee AGJ, Daemen T. Immunologic aspect of ovarian cancer and p53 as tumor antigen. J Transl Med 2005; 3:34. [PMID: 16164749 PMCID: PMC1243238 DOI: 10.1186/1479-5876-3-34] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2005] [Accepted: 09/15/2005] [Indexed: 01/13/2023] Open
Abstract
Ovarian cancer represents the fifth leading cause of death from all cancers for women. During the last decades overall survival has improved due to the use of new chemotherapy schedules. Still, the majority of patients die of this disease. Research reveals that ovarian cancer patients exhibit significant immune responses against their tumor. In this review the knowledge obtained thus far on the interaction of ovarian cancer tumor cells and the immune system is discussed. Furthermore the role of p53 as tumor antigen and its potential role as target antigen in ovarian cancer is summarized. Based on the increased knowledge on the role of the immune system in ovarian cancer major improvements are to be expected of immunotherapy based treatment of this disease.
Collapse
Affiliation(s)
- HW Nijman
- Dept. of Gynaecologic Oncology, Groningen University Medical Center
| | - A Lambeck
- Dept. of Gynaecologic Oncology, Groningen University Medical Center
- Dept. of Medical Microbiology, Molecular Virology Section, Groningen University Medical Center
| | - SH van der Burg
- Dept. of Immunohematology and Blood Transfusion, Leiden University Medical Center
| | - AGJ van der Zee
- Dept. of Gynaecologic Oncology, Groningen University Medical Center
| | - T Daemen
- Dept. of Medical Microbiology, Molecular Virology Section, Groningen University Medical Center
| |
Collapse
|
16
|
Rojas JM, McArdle SEB, Horton RBV, Bell M, Mian S, Li G, Ali SA, Rees RC. Peptide immunisation of HLA-DR-transgenic mice permits the identification of a novel HLA-DRbeta1*0101- and HLA-DRbeta1*0401-restricted epitope from p53. Cancer Immunol Immunother 2005; 54:243-53. [PMID: 15449037 PMCID: PMC11033034 DOI: 10.1007/s00262-004-0596-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2004] [Accepted: 07/07/2004] [Indexed: 10/26/2022]
Abstract
Because of the central role of CD4(+) T cells in antitumour immunity, the identification of the MHC class II-restricted peptides to which CD4(+) T cells respond has become a priority of tumour immunologists. Here, we describe a strategy permitting us to rapidly determine the immunogenicity of candidate HLA-DR-restricted peptides using peptide immunisation of HLA-DR-transgenic mice, followed by assessment of the response in vitro. This strategy was successfully applied to the reported haemaglutinin influenza peptide HA(307-319), and then extended to three candidate HLA-DR-restricted p53 peptides predicted by the evidence-based algorithm SYFPEITHI to bind to HLA-DRbeta1*0101 (HLA-DR1) and HLA-DRbeta1*0401 (HLA-DR4) molecules. One of these peptides, p53(108-122), consistently induced responses in HLA-DR1- and in HLA-DR4-transgenic mice. Moreover, this peptide was naturally processed by dendritic cells (DCs), and induced specific proliferation in the splenocytes of mice immunised with p53 cDNA, demonstrating that immune responses could be naturally mounted to the peptide. Furthermore, p53(108-122) peptide was also immunogenic in HLA-DR1 and HLA-DR4 healthy donors. Thus, the use of this transgenic model permitted the identification of a novel HLA-DR-restricted epitope from p53 and constitutes an attractive approach for the rapid identification of novel immunogenic MHC class II-restricted peptides from tumour antigens, which can ultimately be incorporated in immunotherapeutic protocols.
Collapse
Affiliation(s)
- José Manuel Rojas
- School of Sciences, The Nottingham Trent University, Clifton Lane, Nottingham, NG11 8NS UK
- Department of Haematology, University of Liverpool, Duncan building, Daulby Street, Liverpool, L69 3GA UK
| | | | - Roger B. V. Horton
- School of Sciences, The Nottingham Trent University, Clifton Lane, Nottingham, NG11 8NS UK
| | - Matthew Bell
- School of Sciences, The Nottingham Trent University, Clifton Lane, Nottingham, NG11 8NS UK
| | - Shahid Mian
- School of Sciences, The Nottingham Trent University, Clifton Lane, Nottingham, NG11 8NS UK
| | - Geng Li
- School of Sciences, The Nottingham Trent University, Clifton Lane, Nottingham, NG11 8NS UK
| | - Selman A. Ali
- School of Sciences, The Nottingham Trent University, Clifton Lane, Nottingham, NG11 8NS UK
| | - Robert C. Rees
- School of Sciences, The Nottingham Trent University, Clifton Lane, Nottingham, NG11 8NS UK
| |
Collapse
|
17
|
Abstract
Cutaneous squamous cell carcinoma is typically characterized by the over-expression of the tumour suppressor protein p53. Considerable evidence suggests that immune competence is important in the control of cutaneous SCC. We discuss the immunobiology of p53 and its relevance to cutaneous SCC, including the potential interaction with human papillomavirus.
Collapse
Affiliation(s)
- A P B Black
- University of Oxford, MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, The John Radcliffe Hospital, UK
| | | |
Collapse
|
18
|
Michael A, Pandha HS. Renal-cell carcinoma: tumour markers, T-cell epitopes, and potential for new therapies. Lancet Oncol 2003; 4:215-23. [PMID: 12681265 DOI: 10.1016/s1470-2045(03)01044-1] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Advanced renal-cell carcinoma is a very difficult tumour to treat, and response rates to biological therapies are less than 20%. The identification of various molecular and cellular markers has led to the development of novel therapies. Despite evaluation of their association with histological subtype, immune infiltration, molecular markers of cell proliferation, p53 mutation, and growth-factor-receptor expression, none of these markers has proved better predictive factors than tumour stage and histological grade. The identification of tumour-associated antigens and the specificity of cellular immune responses have led to the development of targeted immunotherapy with monoclonal antibodies, radioimmunotherapy, and T-cell therapies. In this review, we evaluate a range of markers associated with renal-cell carcinoma and new treatment approaches based on tumour-associated antigens and, in particular, T-cell epitopes.
Collapse
|
19
|
Uemura Y, Senju S, Maenaka K, Iwai LK, Fujii S, Tabata H, Tsukamoto H, Hirata S, Chen YZ, Nishimura Y. Systematic analysis of the combinatorial nature of epitopes recognized by TCR leads to identification of mimicry epitopes for glutamic acid decarboxylase 65-specific TCRs. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 170:947-60. [PMID: 12517961 DOI: 10.4049/jimmunol.170.2.947] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Accumulating evidence indicates that recognition by TCRs is far more degenerate than formerly presumed. Cross-recognition of microbial Ags by autoreactive T cells is implicated in the development of autoimmunity, and elucidating the recognition nature of TCRs has great significance for revelation of the disease process. A major drawback of currently used means, including positional scanning synthetic combinatorial peptide libraries, to analyze diversity of epitopes recognized by certain TCRs is that the systematic detection of cross-recognized epitopes considering the combinatorial effect of amino acids within the epitope is difficult. We devised a novel method to resolve this issue and used it to analyze cross-recognition profiles of two glutamic acid decarboxylase 65-autoreactive CD4(+) T cell clones, established from type I diabetes patients. We generated a DNA-based randomized epitope library based on the original glutamic acid decarboxylase epitope using class II-associated invariant chain peptide-substituted invariant chains. The epitope library was composed of seven sublibraries, in which three successive residues within the epitope were randomized simultaneously. Analysis of agonistic epitopes indicates that recognition by both TCRs was significantly affected by combinations of amino acids in the antigenic peptide, although the degree of combinatorial effect differed between the two TCRs. Protein database searching based on the TCR recognition profile proved successful in identifying several microbial and self-protein-derived mimicry epitopes. Some of the identified mimicry epitopes were actually produced from recombinant microbial proteins by APCs to stimulate T cell clones. Our data demonstrate the importance of the combinatorial nature of amino acid residues of epitopes in molecular mimicry.
Collapse
MESH Headings
- Amino Acid Motifs
- Amino Acid Sequence
- Amino Acid Substitution/genetics
- Antigen Presentation/genetics
- Antigens, Differentiation, B-Lymphocyte/genetics
- Autoantigens/analysis
- Autoantigens/metabolism
- Bacterial Proteins/immunology
- Bacterial Proteins/metabolism
- Cells, Cultured
- Clone Cells
- Combinatorial Chemistry Techniques/methods
- Epitopes, T-Lymphocyte/analysis
- Epitopes, T-Lymphocyte/biosynthesis
- Epitopes, T-Lymphocyte/genetics
- Epitopes, T-Lymphocyte/metabolism
- Gene Rearrangement, alpha-Chain T-Cell Antigen Receptor
- Gene Rearrangement, beta-Chain T-Cell Antigen Receptor
- Glutamate Decarboxylase/analysis
- Glutamate Decarboxylase/genetics
- Glutamate Decarboxylase/metabolism
- HLA-DR Antigens/genetics
- HLA-DRB4 Chains
- Histocompatibility Antigens Class II/genetics
- Humans
- Isoenzymes/analysis
- Isoenzymes/genetics
- Isoenzymes/metabolism
- Ligands
- Molecular Mimicry/genetics
- Molecular Mimicry/immunology
- Molecular Sequence Data
- Peptide Library
- Peptides/genetics
- Peptides/immunology
- Peptides/isolation & purification
- Peptides/metabolism
- Receptors, Antigen, T-Cell, alpha-beta/analysis
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
- T-Lymphocyte Subsets/enzymology
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- Viral Proteins/immunology
- Viral Proteins/metabolism
Collapse
Affiliation(s)
- Yasushi Uemura
- Division of Immunogenetics, Department of Neuroscience and Immunology, Kumamoto University Graduate School of Medical Sciences, Kumamoto University School of Medicine, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Makita M, Azuma T, Hamaguchi H, Niiya H, Kojima K, Fujita S, Tanimoto M, Harada M, Yasukawa M. Leukemia-associated fusion proteins, dek-can and bcr-abl, represent immunogenic HLA-DR-restricted epitopes recognized by fusion peptide-specific CD4+ T lymphocytes. Leukemia 2002; 16:2400-7. [PMID: 12454745 DOI: 10.1038/sj.leu.2402742] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2002] [Accepted: 07/23/2002] [Indexed: 11/09/2022]
Abstract
Although CD4(+) helper T lymphocytes have been demonstrated to play an important role in antitumor immune response, only a few epitopes of tumor-associated antigens recognized by HLA class II-restricted CD4(+) T lymphocytes have been identified. In the present study, we addressed the question of whether leukemia-associated fusion proteins are recognized by CD4(+) T lymphocytes. Immature dendritic cells (DCs) were loaded with necrotic or apoptotic leukemia cells with t(6;9) or t(9;22) and then cocultured with the dek-can fusion peptide-specific or the bcr-abl fusion peptide-specific CD4(+) T lymphocyte clone. The dek-can peptide-specific and bcr-abl peptide-specific CD4(+) T lymphocyte clones produced interferon-gamma (IFN-gamma) when they were cocultured with HLA-DR-matched but not with mismatched DCs which had been loaded with apoptotic as well as necrotic leukemia cells with t(6;9) and t(9;22), respectively. IFN-gamma production by CD4(+)T lymphocyte clones in response to stimulation with DCs loaded with leukemia cells was inhibited by the anti-HLA-DR monoclonal antibody. These data indicate that the acute myelogenous leukemia-associated fusion protein, dek-can, and chronic myelogenous leukemia-associated fusion protein, bcr-abl, are both processed and presented by DCs to the fusion peptide-specific CD4(+) T lymphocytes.
Collapse
Affiliation(s)
- M Makita
- First Department of Internal Medicine, Ehime University School of Medicine, Ehime, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Nikitina EY, Chada S, Muro-Cacho C, Fang B, Zhang R, Roth JA, Gabrilovich DI. An effective immunization and cancer treatment with activated dendritic cells transduced with full-length wild-type p53. Gene Ther 2002; 9:345-52. [PMID: 11938454 DOI: 10.1038/sj.gt.3301670] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2001] [Accepted: 12/20/2001] [Indexed: 11/09/2022]
Abstract
P53-based immunization is an attractive approach to cancer immunotherapy due to the accumulation of p53 protein in tumor, but not in normal cells. However, it was not known whether immune response against self-protein (p53) could be generated in vivo. Mouse dendritic cells (DCs) were transduced with adenoviral construct containing murine full-length wild-type p53 (Ad-p53). Repeated immunizations with these cells protected 60% of mice against challenge with MethA sarcoma cells bearing point mutations in p53 gene. Activation of DCs via ligation of CD40 significantly improved the results of immunization: all mice were protected against MethA sarcoma. The treatment of MethA tumor-bearing mice with activated Ad-p53-transduced DCs showed complete tumor rejection in four out of six mice. The specificity of antitumor immune response was confirmed by CTL assay. The analysis of phenotype and function of DCs demonstrated that the effect of CD40 ligation on these cells was enhanced by their infection with Ad-p53. The level of neutralizing anti-adenovirus antibody was moderately elevated in these mice. No signs of autoimmune reaction were evident during detailed pathological evaluation of treated mice. These data demonstrate that activated Ad-p53-infected DCs are able to break tolerance to this protein and can be used in immunotherapy of cancer.
Collapse
Affiliation(s)
- E Y Nikitina
- H Lee Moffitt Cancer Center, University of South Florida, Tampa, FL 33612, USA
| | | | | | | | | | | | | |
Collapse
|
22
|
Tang R, Ko MC, Wang JY, Changchien CR, Chen HH, Chen JS, Hsu KC, Chiang JM, Hsieh LL. Humoral response to p53 in human colorectal tumors: a prospective study of 1,209 patients. Int J Cancer 2001; 94:859-63. [PMID: 11745489 DOI: 10.1002/ijc.1541] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
p53 Antibodies (p53-Abs) have been detected in the serum of a proportion of colorectal cancer (CRC) patients. It is not yet known at which stage during colorectal tumor progression p53-Abs appear in the serum. The utility of these antibodies as markers for CRC prognosis remains to be clarified. Using a quantitative enzyme-linked immunosorbent assay, we analyzed serum samples from 998 CRC patients and from 211 patients with polyp. Levels of p53-Abs were defined as negative (<10 U/microL), low (10-76 U/microL) and high (>76 U/microL). Overall, 13.0% of CRC patients and less than 1% of polyp patients had increased serum p53-Ab levels. High p53-Ab levels were only seen in patients with invasive carcinomas. The parameters that were significantly and independently associated with a greater frequency of high p53-Ab levels were the left colon (odds ratio [OR] = 3.4; 95% CI = 1.1-10.5), the rectum (OR = 2.9; 95% CI, 1.0-8.8) and advanced lymph node metastasis (OR = 4.6; 95% CI, 2.2-9.6). In univariate analysis, patients with high p53-Ab levels had a shorter survival times than did those without (p = 0.007). However, the significant effect disappeared in a Cox regression model adjusting for sex, age, tumor location, carcinoembryonic antigen levels, gross findings, histologic grade, mucin production and TNM stage. Thus, autoantibodies against p53 occur with tumor progression in multistep colorectal carcinogenesis and increase with advanced node metastasis. Furthermore, the seemingly adverse effect of high p53-Ab levels on the survival of CRC patients may be explained by other prognostic factors.
Collapse
Affiliation(s)
- R Tang
- Colorectal Section, Chang Gung Memorial Hospital, Linkou, Taiwan
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Matsushita S, Tanaka Y, Matsuoka T, Nakashima T. Clonal expansion of freshly isolated CD4T cells by randomized peptides and identification of peptide ligands using combinatorial peptide libraries. Eur J Immunol 2001; 31:2395-402. [PMID: 11500823 DOI: 10.1002/1521-4141(200108)31:8<2395::aid-immu2395>3.0.co;2-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
We synthesized Xn (n = 9 -- 19) peptides that consist of 9 to 19 residues with random sequences. X19 is considered to deliver antigenic stimuli to CD4 T cells, because: (a) X19 induces proliferation of peripheral blood mononuclear cells (PBMC), in the presence of IL-2, which is abrogated by monoclonal antibodies to class II HLA; (b) X19 + IL-2 induces proliferation of CD4 T cell clones of distinct specificities; and (c) T cell clones recognizing the same TCR ligands with distinct V beta usage are equally stimulated by X19 + IL-2. We next co-cultured single peripheral CD4 T cells with X19 and mitomycin-treated autologous PBMC. Indeed, single T cells of CD45RA(-) memory phenotype exhibited clonal expansion, with variable rates of proliferation, when IL-4, IL-7, IL-9, IL-15 and agonistic antibody to CD29 were included in the culture. These T cell clones showed heterogeneous proliferation patterns against KGXXXXXXXXXGK-based and KGXXXXXXXXXGKGKK-based combinatorial peptides libraries, in the presence of IL-2. Pattern-match search on a T cell clone resulted in peptide ligand candidates, one of which induced proliferation, as did protein molecules carrying the corresponding sequence. These results indicate that X19 can induce proliferation of peripheral memory T cells, the peptide ligands of which can be determined using combinatorial peptide libraries.
Collapse
Affiliation(s)
- S Matsushita
- Division of Immunogenetics, Department of Neuroscience and Immunology, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan.
| | | | | | | |
Collapse
|
24
|
Ferriès E, Connan F, Pagès F, Gaston J, Hagnéré AM, Vieillefond A, Thiounn N, Guillet J, Choppin J. Identification of p53 peptides recognized by CD8(+) T lymphocytes from patients with bladder cancer. Hum Immunol 2001; 62:791-8. [PMID: 11476902 DOI: 10.1016/s0198-8859(01)00266-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
In many types of cancer, p53 frequently accumulates in tumor cells and anti-p53 antibodies can be detected. However, only four CD8(+) T-cell epitopes from p53 have been identified in humans so far. To further analyze the development of a T-cell response against p53, peptides having binding motifs specific for HLA-A1, -A2, -A3, -A24, -B7, -B35, -B44, and -B51 molecules have been defined. The HLA-binding capacity of those peptides was tested, and the stability of formed complexes was defined. Thirteen peptides that bound to HLA-A24 and -B44 molecules are presented. The positive peptides were then used to detect the anti-p53 response of CD8(+) T lymphocytes from patients with bladder cancer. Six peptides, presented by HLA-A2, -B51, or -A24, were able to stimulate T cells from two patients (among 16) with tumor cells that strongly accumulated p53. On the contrary, p53 peptides systematically failed to stimulate T cells from healthy donors or patients with low or undetectable levels of p53 in their tumor cells. These results have led to the identification of four new potential T CD8(+) epitopes from p53: 194-203 associating with HLA-B51 and 204-212, 211-218, and 235-243 associating with HLA-A24.
Collapse
Affiliation(s)
- E Ferriès
- INSERM U445, Laboratoire Associé No9 du Comité de Paris de la Ligue contre le Cancer, Institut Cochin de Génétique Moléculaire, Université René Descartes, Paris, France.
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Matsuoka T, Tabata H, Matsushita S. Monocytes are differentially activated through HLA-DR, -DQ, and -DP molecules via mitogen-activated protein kinases. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 166:2202-8. [PMID: 11160273 DOI: 10.4049/jimmunol.166.4.2202] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
When HLA-DR, -DQ, and -DP were cross-linked by solid-phase mAbs, monocytes produced monokines and only anti-DR markedly activated mitogen-activated protein (MAP) kinase extracellular signal-related kinase, whereas anti-DR, anti-DQ, and anti-DP all activated MAP kinase p38. Activation of extracellular signal-related kinase was not inhibited by neutralizing Ab to TNF-alpha. Anti-DR and DR-restricted T cells stimulated monocytes to produce relatively higher levels of proinflammatory monokines, such as IL-1beta, whereas anti-DQ/DP and DQ-/DP-restricted T cells stimulated higher levels of anti-inflammatory monokine IL-10. IL-10 production was abrogated by the p38 inhibitor SB203580, but rather enhanced by the MAP/extracellular signal-related kinase kinase-I-specific inhibitor PD98059, whereas IL-1beta was only partially abrogated by SB203580 and PD98059. Furthermore, DR-restricted T cells established from PBMC, which are reactive with mite Ags, purified protein derivative, and random 19-mer peptides, exhibited a higher IFN-gamma:IL-4 ratio than did DQ- or DP-restricted T cells. These results indicate that HLA-DR, -DQ, and -DP molecules transmit distinct signals to monocytes via MAP kinases and lead to distinct monokine activation patterns, which may affect T cell responses in vivo. Thus, the need for generation of a multigene family of class II MHC seems apparent.
Collapse
Affiliation(s)
- T Matsuoka
- Department of Neuroscience and Immunology, Division of Immunogenetics, Kumamoto University Graduate School of Medical Sciences, Honjo, Kumamoto, Japan
| | | | | |
Collapse
|
26
|
Rosenwirth B, Kuhn EM, Heeney JL, Hurpin C, Tartaglia J, Bonnet MC, Moingeon P, Erdile L. Safety and immunogenicity of ALVAC wild-type human p53 (vCP207) by the intravenous route in rhesus macaques. Vaccine 2001; 19:1661-70. [PMID: 11166889 DOI: 10.1016/s0264-410x(00)00416-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
p53 is over-expressed in approximately 50% of human cancers, and transfer of cytotoxic T lymphocytes (CTL) against wild-type p53 protects mice against p53-over-expressing tumors, suggesting that p53 might be an attractive target for immunotherapy. Immunization of mice with a recombinant canarypox virus, ALVAC, expressing human wild-type p53 (vCP207) prevented growth of p53-over-expressing tumors. Since intravenous administration induced better immune responses in mice than other routes, we have proposed to use this route in cancer patients. However, because this vector has never been administered intravenously to humans, and because of the possibility of inducing auto-immunity to a self-antigen, we felt it was necessary to first evaluate safety in rhesus macaques. We found that three intravenous administrations of vCP207 at proportional doses up to 10x those proposed for humans produced no abnormalities in hematologic or clinical chemistry parameters. Serologic markers of autoimmunity and inflammation were unaffected, despite the >95% amino acid identity between human and rhesus p53. Pathological examination of numerous tissues yielded findings comparable to those in animals given placebo. Some animals showed anti-p53 antibody responses following vaccination, indicating that tolerance could be broken to some extent. However, with the exception of one animal with a possible delayed type hypersensitivity reaction to p53 protein, we did not see evidence for a cell-mediated response. The safety profile in monkeys with ALVAC-p53 provides encouragement for using such live, modified vectors via the intravenous route for human immunotherapy.
Collapse
Affiliation(s)
- B Rosenwirth
- Biomedical Primate Research Centre, Rijswijk, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
27
|
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: 45] [Impact Index Per Article: 2.0] [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
|
28
|
Seliger B, Maeurer MJ, Ferrone S. Antigen-processing machinery breakdown and tumor growth. IMMUNOLOGY TODAY 2000; 21:455-64. [PMID: 10953098 DOI: 10.1016/s0167-5699(00)01692-3] [Citation(s) in RCA: 317] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Defects in the major histocompatibility complex (MHC) class I antigen-processing machinery (APM) have been described in tumors of different histology. Murine data suggest that defects in the MHC class II APM might also be associated with malignant transformation of human cells. This article describes the pathophysiology of the MHC class I and II APM, reviews APM abnormalities in tumor cells and discusses their role in the escape of tumor cells from in vitro recognition by T cells.
Collapse
Affiliation(s)
- B Seliger
- Johannes Gutenberg-University, III. Dept of Internal Medicine, Langenbeckstr. 1, 55131 Mainz, Germany.
| | | | | |
Collapse
|
29
|
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: 1.8] [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
|
30
|
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
|
31
|
O'Byrne KJ, Dalgleish AG, Browning MJ, Steward WP, Harris AL. The relationship between angiogenesis and the immune response in carcinogenesis and the progression of malignant disease. Eur J Cancer 2000; 36:151-69. [PMID: 10741273 DOI: 10.1016/s0959-8049(99)00241-5] [Citation(s) in RCA: 128] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Recent studies have demonstrated that angiogenesis and suppressed cell-mediated immunity (CMI) play a central role in the pathogenesis of malignant disease facilitating tumour growth, invasion and metastasis. In the majority of tumours, the malignant process is preceded by a pathological condition or exposure to an irritant which itself is associated with the induction of angiogenesis and/or suppressed CMI. These include: cigarette smoking, chronic bronchitis and lung cancer; chronic oesophagitis and oesophageal cancer; chronic viral infections such as human papilloma virus and ano-genital cancers, chronic hepatitis B and C and hepatocellular carcinoma, and Epstein-Barr virus (EBV) and lymphomas; chronic inflammatory conditions such as Crohn's disease and ulcerative colitis and colorectal cancer; asbestos exposure and mesothelioma and excessive sunlight exposure/sunburn and malignant melanoma. Chronic exposure to growth factors (insulin-like growth factor-I in acromegaly), mutations in tumour suppressor genes (TP53 in Li Fraumeni syndrome) and long-term exposure to immunosuppressive agents (cyclosporin A) may also give rise to similar environments and are associated with the development of a range of solid tumours. The increased blood supply would facilitate the development and proliferation of an abnormal clone or clones of cells arising as the result of: (a) an inherited genetic abnormality; and/or (b) acquired somatic mutations, the latter due to local production and/or enhanced delivery of carcinogens and mutagenic growth factors. With progressive detrimental mutations and growth-induced tumour hypoxia, the transformed cell, to a lesser or greater extent, may amplify the angiogenic process and CMI suppression, thereby facilitating further tumour growth and metastasis. There is accumulating evidence that long-term treatment with cyclo-oxygenase inhibitors (aspirin and indomethacin), cytokines such as interferon-alpha, anti-oestrogens (tamoxifen and raloxifene) and captopril significantly reduces the incidence of solid tumours such as breast and colorectal cancer. These agents are anti-angiogenic and, in the case of aspirin, indomethacin and interferon-alpha have proven immunomodulatory effects. Collectively these observations indicate that angiogenesis and suppressed CMI play a central role in the development and progression of malignant disease.
Collapse
Affiliation(s)
- K J O'Byrne
- University Department of Oncology, Leicester Royal Infirmary, UK.
| | | | | | | | | |
Collapse
|
32
|
Tanaka Y, Ohyama H, Ogawa M, Nishimura Y, Matsushita S. Identification of Peptide Superagonists for a Self-K-ras-Reactive CD4+ T Cell Clone Using Combinatorial Peptide Libraries and Mass Spectrometry. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.162.12.7155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Abstract
The proliferative responses of a human CD4+ T cell clone 29.15.2, reactive with a self-K-ras-derived peptide (3EYKLVVVGAGGVGKSALT20), were tested using a set of X9 combinatorial peptide libraries containing the flanking residues (EYKLVXXXXXXXXXSALT, where X indicates random amino acids). Certain peptide libraries, such as EYKLVXXXXXXMXXSALT and EYKLVXXXXXXXHXSALT, stimulated a marked proliferation of 29.15.2. However, no combinations of substitutions tested, such as EYKLVXXXXXXMHXSALT, exhibited additive effects. We subsequently synthesized peptides with degenerate sequences (a mixture of 480 species), where each position is composed of the wild-type (wt) residue or of amino acids that induced the proliferation of 29.15.2, in positional scanning. Interestingly, one fraction of degenerate peptides, separated by reverse-phase HPLC, stimulated much higher proliferation than did the wt; in addition, the retention time of this fraction was distinct from that of the wt. Mass spectrometry analysis of this fraction and flanking fractions identified five peptide species that exhibit strong signals in a manner that parallels the antigenic activity. Finally, 17 candidate peptide sequences were deduced from mass spectrometry and hydrophobicity scoring results, of which two peptides (EYKLVVVGAGGMLKSALT and EYKLVVVGAGGMIKSALT) did induce 52- and 61-fold stronger proliferation, respectively, compared with the wt. These findings indicate that: 1) synthetic peptides that carry “the best” residue substitution at each position of combinatorial peptide libraries do not always exhibit superagonism, and 2) such a drawback can be overcome with the use of mass spectrometry. This approach provides new perspectives for the accurate and efficient identification of peptide superagonists.
Collapse
Affiliation(s)
- Yoshihiko Tanaka
- *Division of Immunogenetics, Department of Neuroscience and Immunology, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
- †Department of Surgery II, Kumamoto University School of Medicine, Kumamoto, Japan; and
| | - Hideki Ohyama
- ‡Department of Periodontology and Endodontology, Okayama University Dental School, Okayama, Japan
| | - Michio Ogawa
- †Department of Surgery II, Kumamoto University School of Medicine, Kumamoto, Japan; and
| | - Yasuharu Nishimura
- *Division of Immunogenetics, Department of Neuroscience and Immunology, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
| | - Sho Matsushita
- *Division of Immunogenetics, Department of Neuroscience and Immunology, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
| |
Collapse
|
33
|
Tabata H, Kanai T, Yoshizumi H, Nishiyama S, Fujimoto S, Matsuda I, Yasukawa M, Matsushita S, Nishimura Y. Characterization of self-glutamic acid decarboxylase 65-reactive CD4+ T-cell clones established from Japanese patients with insulin-dependent diabetes mellitus. Hum Immunol 1998; 59:549-60. [PMID: 9757911 DOI: 10.1016/s0198-8859(98)00050-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
To investigate autoimmunity to glutamic acid decarboxylase (GAD) 65 in Japanese patients with insulin-dependent diabetes mellitus (IDDM, type I diabetes), we established seven CD4+ T-cell clones, by stimulating peripheral blood mononuclear cells (PBMC) of six IDDM patients, using a mixture of overlapping human GAD65 peptides. No GAD65 autoreactive T-cell clones were evidenced in four healthy controls. Specificities of T-cell clones were as follows: (a) two clones specific to GAD65 p111-131 (residue 111 to 131) + DR53 (DRB4*0103); (b) one clone specific to GAD65 p413-433 + DR1 (DRB1*0101); (c) two clones specific to GAD65 p200-217 + either DR9 (DRB1*0901) or DR8 (DRB1*0802); and (d) two clones specific to GAD65 p368-388 + DP2 (DPA1*01 or 0201-DPB1*0201). Two DR53-restricted and one DR1-restricted T-cell clones, responded to a recombinant human GAD65 protein, and showed cytotoxicity against B lymphoblastoid cell lines pre-pulsed with the peptides. Six T-cell clones exhibited the Th1-like phenotype. Interestingly, two DR53-restricted T-cell clones killed a Fas-deficient B lymphoblastoid cell line, thereby indicating that cytotoxicity was not completely dependent on a Fas-Fas ligand interaction. Thus, the T-cell epitopes were mapped in a limited portion of GAD65 protein, with a tendency to be restricted by disease-associated HLA-DR, but not DQ molecules.
Collapse
Affiliation(s)
- H Tabata
- Department of Neuroscience and Immunology, Kumamoto University Graduate School of Medical Sciences, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|