351
|
Cheng WF, Hung CF, Lin KY, Ling M, Juang J, He L, Lin CT, Wu TC. CD8+ T cells, NK cells and IFN-gamma are important for control of tumor with downregulated MHC class I expression by DNA vaccination. Gene Ther 2003; 10:1311-20. [PMID: 12883527 DOI: 10.1038/sj.gt.3301982] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
One of the major hurdles facing cancer immunotherapy is that cancers may downregulate expression of MHC class I molecules. The development of a suitable tumor model with downregulated MHC class I expression is critical for designing vaccines and immunotherapeutic strategies to control such tumors. We developed an E7-expressing murine tumor model with downregulated MHC class I expression, TC-1 P3 (A15). Using this model, we tested DNA and vaccinia vaccines for their ability to control tumors with downregulated MHC class I expression. We found that vaccination with DNA encoding E7 linked to Mycobacterial heat shock protein 70 (HSP70) generated a significant antitumor effect against TC-1 P3 (A15), while vaccination with E7/HSP70 vaccinia did not generate an appreciable antitumor effect. Lymphocyte depletion experiments revealed that both CD8+ T cells and NK cells were essential for the antitumor effect generated by E7/HSP70 DNA against TC-1 P3 (A15). Furthermore, tumor protection experiments using IFN-gamma knockout mice revealed that IFN-gamma was essential for the antitumor effect generated by E7/HSP70 DNA against TC-1 P3 (A15). Our results demonstrate that vaccination with E7/HSP70 DNA results in a significant antitumor effect against a neoplasm with downregulated MHC class I expression and the importance of CD8+ T cells, NK cells, and IFN-gamma in generating this antitumor effect.
Collapse
Affiliation(s)
- W F Cheng
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | | | | | | | | | | | | | | |
Collapse
|
352
|
Da Silva DM, Schiller JT, Kast WM. Heterologous boosting increases immunogenicity of chimeric papillomavirus virus-like particle vaccines. Vaccine 2003; 21:3219-27. [PMID: 12804851 DOI: 10.1016/s0264-410x(03)00237-8] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Chimeric human papillomavirus virus-like particles (HPV cVLPs), containing the HPV16 non-structural protein E7, are potent vaccines for inducing antigen-specific protective immunity against HPV-transformed tumors in animal models. Previous data demonstrated that the effectiveness of cytotoxic T lymphocyte (CTL) induction after repetitive vaccination with the same cVLP, and thus vaccine efficacy, is limited by the presence of neutralizing antibodies induced after the first application. Here, we determined if altering the route of vaccine delivery or incorporation of the target antigen into VLPs of a heterologous papillomavirus type could overcome inhibition of MHC class I antigen presentation by neutralizing antibodies, resulting in a boosting of CD8(+) T-cell responses against the incorporated antigen, HPV16 E7. Mucosal delivery of cVLPs resulted in detection of systemic E7-specific CD8(+) T cells, however, these routes were not able to bypass the inhibitory effect of circulating antibodies against homologous VLP types. In contrast, mice immunized and boosted with heterologous cVLPs containing HPV16 E7 showed a higher frequency of E7-specific T cells in vitro and displayed reduced tumor growth in a therapeutic setting compared to mice treated with homologous cVLPs. The data indicate that the use of different cVLP types for prime/boost regimens is a promising strategy to increase the efficacy and usefulness of cVLP-based vaccines for the treatment of cervical neoplasia.
Collapse
Affiliation(s)
- Diane M Da Silva
- Cancer Immunology Program, Department of Microbiology and Immunology, Cardinal Bernardin Cancer Center, Loyola University Chicago, 2160 S. First Avenue, Maywood, IL, 60153, USA
| | | | | |
Collapse
|
353
|
Kotecha MT, Afghan RK, Vasilikopoulou E, Wilson E, Marsh P, Kast WM, Davies DH, Caparros-Wanderley W. Enhanced tumour growth after DNA vaccination against human papilloma virus E7 oncoprotein: evidence for tumour-induced immune deviation. Vaccine 2003; 21:2506-15. [PMID: 12744885 DOI: 10.1016/s0264-410x(03)00043-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
We have examined the induction of anti-tumour immunity in a murine model using a gene vaccine approach to deliver a well defined tumour antigen. The vaccines expressed the human papilloma virus type 16 (HPV 16) E7 oncoprotein, and protection was measured against HPV 16-expressing C3R tumour cell line in vivo. In control mice injected with saline, C3R cells initially formed tumours but then regressed completely. As expected, animals injected with a peptide that represents the D(b)-presented CTL epitope from E7 (RAHYNIVTF) were completely protected from tumour growth. Contrary to expectation, however, we consistently saw enhanced tumour growth, delayed regression, or tumour outgrowth in mice vaccinated with two different E7-expressing DNA vaccines. We found no evidence for loss of D(b) or K(b) class I MHC molecules from C3R cells recovered from outgrown tumours, and fluorescent MHC/peptide tetramer staining revealed E7 gene vaccination did not delete RAHYNIVTF-specific CD8(+) T cells. However, we did observe an effect on cytokine production. Splenocytes from E7 gene vaccinated animals responded to re-stimulation in vitro with C3R cells by producing IL-4 but background levels of IFN-gamma. We also observed that cytokine production and E7 peptide-specific CTL were only detectable in vaccinated animals after C3R challenge, but not after DNA priming alone. We conclude that 'prime-boosting' is necessary to observe tumour-specific T cell responses with the gene vaccine approach, but that boosting with tumour cells causes skewing of the primed cells in a T2 direction that is incompatible with protective anti-tumour immunity.
Collapse
Affiliation(s)
- Minal T Kotecha
- Infection & Immunity Research Group, School of Health & Life Sciences, King's College London, London, UK
| | | | | | | | | | | | | | | |
Collapse
|
354
|
Dileo J, Banerjee R, Whitmore M, Nayak JV, Falo LD, Huang L. Lipid-protamine-DNA-mediated antigen delivery to antigen-presenting cells results in enhanced anti-tumor immune responses. Mol Ther 2003; 7:640-8. [PMID: 12718907 DOI: 10.1016/s1525-0016(03)00064-9] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
Vaccination with antigenic peptides encoding tumor antigens has the potential to be an effective treatment for cancer. To induce tumor-specific cellular immune responses, a peptide antigen must be presented by antigen-presenting cells (APCs) to T-cells in the lymphatic tissues. Effective in vivo delivery of peptide antigens to APCs has been problematic. Here we use a model antigen from the HPV16 E7 protein to formulate LPD/E7 particles that upon iv administration are internalized by CD11c(+) and CD11b(+) cells in the marginal zone of the spleen. Either iv or sc vaccination with LPD/E7 particles induces E7-specific CTL responses stronger than those obtained using previously described liposome/peptide strategies and prevents the establishment of E7-expressing tumors. Furthermore, the administration of LPD/E7 particles to tumor-bearing mice caused complete tumor regression in 100% of the treated animals. Based on these studies, the entrapment of peptide antigens inside LPD particles may be an effective and generally applicable strategy for the enhancement of peptide vaccine potency.
Collapse
Affiliation(s)
- John Dileo
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, Pittsburgh, Pennsylvania 15213, USA
| | | | | | | | | | | |
Collapse
|
355
|
de Jong A, O'Neill T, Khan AY, Kwappenberg KMC, Chisholm SE, Whittle NR, Dobson JA, Jack LC, St Clair Roberts JA, Offringa R, van der Burg SH, Hickling JK. Enhancement of human papillomavirus (HPV) type 16 E6 and E7-specific T-cell immunity in healthy volunteers through vaccination with TA-CIN, an HPV16 L2E7E6 fusion protein vaccine. Vaccine 2003; 20:3456-64. [PMID: 12297390 DOI: 10.1016/s0264-410x(02)00350-x] [Citation(s) in RCA: 128] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
TA-CIN is a vaccine that comprises the human papillomavirus (HPV) type 16 L2, E6 and E7 as a single fusion protein. In a mouse model, TA-CIN effectively prevented outgrowth of HPV16-positive tumour cells. To assess the safety and immunogenicity of TA-CIN, a dose escalating (26, 128, 533 micro g), double blind and placebo-controlled phase I study was conducted in 40 healthy volunteers. TA-CIN was administered without adjuvant by intramuscular injection on weeks 0, 4 and 8. No serious adverse events of the vaccination were reported during the study. Both IgG antibodies and proliferative responses against TA-CIN were elicited at all three doses. More importantly, T-cell immunity against the HPV16 E6 and E7 oncoproteins was detected by IFN gamma ELISPOT in 8/11 evaluable subjects vaccinated with the 533 micro g dose.
Collapse
Affiliation(s)
- A de Jong
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Centre, PO Box 9600, 2300 RC, Leiden, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
356
|
Eiben GL, Velders MP, Kast WM. The cell-mediated immune response to human papillomavirus-induced cervical cancer: implications for immunotherapy. Adv Cancer Res 2003; 86:113-48. [PMID: 12374277 DOI: 10.1016/s0065-230x(02)86004-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Gretchen L Eiben
- Cardinal Bernardin Cancer Center, Loyola University Chicago, Maywood Illinois 60153, USA
| | | | | |
Collapse
|
357
|
Ohlschläger P, Osen W, Dell K, Faath S, Garcea RL, Jochmus I, Müller M, Pawlita M, Schäfer K, Sehr P, Staib C, Sutter G, Gissmann L. Human papillomavirus type 16 L1 capsomeres induce L1-specific cytotoxic T lymphocytes and tumor regression in C57BL/6 mice. J Virol 2003; 77:4635-45. [PMID: 12663770 PMCID: PMC152157 DOI: 10.1128/jvi.77.8.4635-4645.2003] [Citation(s) in RCA: 81] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We analyzed capsomeres of human papillomavirus type 16 (HPV16) consisting of the L1 major structural protein for their ability to trigger a cytotoxic T-cell (CTL) response. To this end, we immunized C57BL/6 mice and used the L1(165-173) peptide for ex vivo restimulation of splenocytes prior to analysis ((51)Cr release assay and enzyme-linked immunospot assay [ELISPOT]). This peptide was identified in this study as a D(b)-restricted naturally processed CTL epitope by HPV16 L1 sequence analysis, major histocompatibility complex class I binding, and (51)Cr release assays following immunization of C57BL/6 mice with HPV16 L1 virus-like particles (VLPs). HPV16 L1 capsomeres were obtained by purification of HPV16 L1 lacking 10 N-terminal amino acids after expression in Escherichia coli as a glutathione S-transferase fusion protein (GST-HPV16 L1 Delta N10). Sedimentation analysis revealed that the majority of the purified protein consisted of pentameric capsomeres, and assembled particles were not observed in minor contaminating higher-molecular-weight material. Subcutaneous (s.c.) as well as intranasal immunization of C57BL/6 mice with HPV16 L1 capsomeres triggered an L1-specific CTL response in a dose-dependent manner as measured by ELISPOT and (51)Cr release assay. Significant reduction of contaminating bacterial endotoxin (lipopolysaccharide) from the capsomere preparation did not diminish the immunogenicity. Antibody responses (serum and vaginal) were less robust under the experimental conditions employed. In addition, s.c. vaccination with HPV16 L1 capsomeres induced regression of established tumors expressing L1 determinants (C3 tumor cells). Our data demonstrate that capsomeres are potent inducers of CTL responses similar to completely assembled T=7 VLPs. This result is of potential relevance for the development of (combined prophylactic and therapeutic) HPV-specific vaccines, since capsomeres can be produced easily and also can be modified to incorporate heterologous sequences such as early HPV proteins.
Collapse
Affiliation(s)
- Peter Ohlschläger
- Angewandte Tumorvirologie, Deutsches Krebsforschungszentrum, D-69120 Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
358
|
Wiethe C, Dittmar K, Doan T, Lindenmaier W, Tindle R. Provision of 4-1BB ligand enhances effector and memory CTL responses generated by immunization with dendritic cells expressing a human tumor-associated antigen. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 170:2912-22. [PMID: 12626542 DOI: 10.4049/jimmunol.170.6.2912] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Up-regulation of receptor-ligand pairs during interaction of an MHC-presented epitope on dendritic cells (DCs) with cognate TCR may amplify, sustain, and drive diversity in the ensuing T cell immune response. Members of the TNF ligand superfamily and the TNFR superfamily contribute to this costimulatory molecule signaling. In this study, we used replication deficient adenoviruses to introduce a model tumor-associated Ag (the E7 oncoprotein of human papillomavirus 16) and the T cell costimulatory molecule 4-1BBL into murine DCs, and monitored the ability of these recombinant DCs to elicit E7-directed T cell responses following immunization. Splenocytes from mice immunized with DCs expressing E7 alone elicited E7-directed effector and memory CTL responses. Coexpression of 4-1BBL in these E7-expressing DCs increased effector and memory CTL responses when they were used for immunization. 4-1BBL expression up-regulated CD80 and CD86 second signaling molecules in DCs. We also report an additive effect of 4-1BBL and receptor activator of NF-kappa B/receptor activator of NF-kappa B ligand coexpression in E7-transduced DC immunogens on E7-directed effector and memory CTL responses and on MHC class II and CD80/86 expression in DCs. Additionally, expression of 4-1BBL in E7-transduced DCs reduced nonspecific T cell activation characteristic of adenovirus vector-associated immunization. The results have generic implications for improved or tumor Ag-expressing DC vaccines by incorporation of exogenous 4-1BBL. There are also specific implications for an improved DC-based vaccine for human papillomavirus 16-associated cervical carcinoma.
Collapse
MESH Headings
- 4-1BB Ligand
- Adenovirus E1A Proteins/biosynthesis
- Adenovirus E1A Proteins/genetics
- Adenovirus E1A Proteins/physiology
- Adenoviruses, Human/genetics
- Adenoviruses, Human/immunology
- Adjuvants, Immunologic/biosynthesis
- Adjuvants, Immunologic/genetics
- Adjuvants, Immunologic/physiology
- Amino Acid Sequence
- Animals
- Antigens, CD/biosynthesis
- Antigens, Neoplasm/administration & dosage
- Antigens, Neoplasm/immunology
- B7-1 Antigen/biosynthesis
- B7-2 Antigen
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/immunology
- Carrier Proteins/biosynthesis
- Carrier Proteins/genetics
- Cell Count
- Cell Line, Transformed
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Dendritic Cells/transplantation
- Dendritic Cells/virology
- Genetic Vectors
- Glycoproteins/biosynthesis
- Glycoproteins/genetics
- Histocompatibility Antigens Class II/biosynthesis
- Humans
- Immunologic Memory/genetics
- Ligands
- Lymphocyte Activation/genetics
- Membrane Glycoproteins/biosynthesis
- Membrane Glycoproteins/genetics
- Mice
- Mice, Inbred A
- Mice, Inbred C57BL
- Molecular Sequence Data
- Osteoprotegerin
- RANK Ligand
- Receptor Activator of Nuclear Factor-kappa B
- Receptors, Cytoplasmic and Nuclear/biosynthesis
- Receptors, Cytoplasmic and Nuclear/genetics
- Receptors, Tumor Necrosis Factor
- Recombination, Genetic
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/virology
- Transduction, Genetic
- Tumor Cells, Cultured
- Tumor Necrosis Factor-alpha/biosynthesis
- Tumor Necrosis Factor-alpha/genetics
- Tumor Necrosis Factor-alpha/physiology
- Up-Regulation/genetics
- Up-Regulation/immunology
Collapse
Affiliation(s)
- Carsten Wiethe
- Department of Molecular Biotechnology, Gesellschaft für Biotechnologische Forschung, Braunschweig, Germany
| | | | | | | | | |
Collapse
|
359
|
Daemen T, Riezebos-Brilman A, Bungener L, Regts J, Dontje B, Wilschut J. Eradication of established HPV16-transformed tumours after immunisation with recombinant Semliki Forest virus expressing a fusion protein of E6 and E7. Vaccine 2003; 21:1082-8. [PMID: 12559783 DOI: 10.1016/s0264-410x(02)00558-3] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Previously, we described the efficacy of immunisation with recombinant Semliki Forest virus (SFV), expressing the human papillomavirus 16 (HPV) oncoproteins E6 and E7, in inducing HPV-specific CTLs and anti-tumour responses. Recently, we developed a novel recombinant SFV construct encoding a relatively stable fusion protein of HPV16 E6 and E7 under control of a translational enhancer derived from the SFV capsid protein. In the present study we demonstrate that immunisation of tumour-bearing mice with this improved vector results in the regression and complete elimination of established tumours. We furthermore demonstrate that a long-term high level of CTL activity, up to 340 days, accompanies the anti-tumour response. Thus, immunisation with recombinant SFV particles encoding increased levels of a fusion protein of HPV16 E6 and E7 efficiently induces CTL activity and CTL memory resulting in a potent therapeutic anti-tumour effect.
Collapse
Affiliation(s)
- Toos Daemen
- Department of Medical Microbiology, Molecular Virology Section, University of Groningen, Groningen, The Netherlands.
| | | | | | | | | | | |
Collapse
|
360
|
Gey A, Kumari P, Sambandam A, Lecuru F, Cassard L, Badoual C, Fridman C, Nagarajan B, Fridman WH, Tartour E. Identification and characterisation of a group of cervical carcinoma patients with profound downregulation of intratumoral Type 1 (IFNgamma) and Type 2 (IL-4) cytokine mRNA expression. Eur J Cancer 2003; 39:595-603. [PMID: 12628838 DOI: 10.1016/s0959-8049(02)00839-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Type 1 cytokines, such as interferon gamma (IFNgamma) and interleukin-2 (IL-2), increase T cell-mediated immune responses and are considered to be beneficial for antitumour immunity. Type 2 cytokines, such as IL-4, IL-5, and IL-10, inhibit Type 1 responses and promote humoral responses. We have previously reported an association between low intratumoral IFNgamma mRNA levels and poor clinical outcome in patients with invasive cervical carcinoma. In this study, by using quantitative polymerase chain reaction (PCR), we identified a group of cervical carcinoma patients with undetectable intratumoral T cell-derived cytokine mRNAs, as IFNgamma, IL-4 and IL-17 expression could not be detected in 5, 25 and 8 of the 52 biopsies analysed, respectively. Global downregulation of Type 1 and Type 2 cytokines was observed in a subgroup of patients who more frequently presented advanced stage tumours. Biopsies of patients with no IFNgamma gene expression did not appear to be less infiltrated by T cells than control biopsies with measurable IFNgamma gene expression. These results clearly demonstrate that, in some clinical situations, the decrease in intratumoral Type 1 cytokines is not associated with a Type 2 polarisation, but rather reflects global deactivation of T cells at the tumour site. These data provide support for immunotherapy protocols designed to reverse the anergic state of T cells in cancer.
Collapse
Affiliation(s)
- A Gey
- Unité d'immunologie biologique, Hopital Européen Georges Pompidou, INSERM U255, Université Pierre et Marie Curie, 20 Rue Leblanc 75908, Paris Cedex 15, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
361
|
Abstract
There is a renewed enthusiasm for therapeutic vaccination as a viable treatment for patients with cancer. Early tumor vaccines were comprised of whole tumor cells, fragments of tumor cells, or protein lysate from tumor cells. Limited results with these approaches led investigators to begin developing the next generation of cancer vaccines based on defined tumor-associated antigens (TAAs). Defining and characterizing TAAs for human cancer, development of new approaches for identifying TAAs, and novel strategies to deliver the antigens as potent therapeutic vaccines have all been the focus of intense research in the past decade and will continue to be the focus for decades to come.
Collapse
Affiliation(s)
- Jennifer D Lewis
- Department of Microbiology and Immunology and the Southwest Cancer Center, Texas Tech University Health Sciences Center, Lubbock, Texas 79430, USA
| | | | | |
Collapse
|
362
|
Dols A, Meijer SL, Smith JW, Fox BA, Urba WJ. Allogeneic breast cancer cell vaccines. Clin Breast Cancer 2003; 3 Suppl 4:S173-80. [PMID: 12620156 DOI: 10.3816/cbc.2003.s.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Cancer vaccines are currently a major focus of immunotherapy research. The combination of specific targeting and low levels of toxicity makes vaccination an attractive approach. There are a variety of immunogens that can be employed to vaccinate patients in order to induce or enhance an antitumor response. The observation that most T-cell priming occurs via presentation of tumor antigens from tumor cells engulfed by host antigen-presenting cells, rather than by direct presentation by vaccine tumor cells themselves, provides the immunological rationale for an allogeneic tumor cell vaccine approach. Furthermore, there are practical advantages over an autologous tumor cell vaccine approach. We summarize herein the limited experience using allogeneic whole cell vaccines in patients with breast cancer. We also describe in vitro immunological results using peripheral blood mononuclear cells from women with stage IV breast cancer who were enrolled in a phase I trial employing a human leukocyte antigen-A2-matched, CD80-modified, allogeneic, whole cell vaccine. Clinical trials employing allogeneic tumor cell vaccines have achieved encouraging immunological and clinical effects in stage IV patients. Allogeneic tumor cell vaccines are safe, feasible, and associated with low toxicity, and the early clinical results suggest that they are worthy of further study
Collapse
Affiliation(s)
- Annemieke Dols
- Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute, Providence Portland Medical Center, OR 97213, USA
| | | | | | | | | |
Collapse
|
363
|
Khammanivong V, Liu XS, Liu WJ, Rodda SJ, Leggatt GR, Tindle RW, Frazer IH, Fernando GJP. Paucity of functional CTL epitopes in the E7 oncoprotein of cervical cancer associated human papillomavirus type 16. Immunol Cell Biol 2003; 81:1-7. [PMID: 12534940 DOI: 10.1046/j.1440-1711.2003.01130.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: 02/01/2023]
Abstract
Many specific antiviral and antitumour immune responses have been attributed to the protective effects of antigen-specific CD8+ cytotoxic T lymphocytes (CTL). Recognition of virus infected or tumour cells by CTL requires presentation of at least one peptide epitope from a virus or tumour-specific antigen by the relevant MHC Class I molecule. Viral genes with mutations which remove CTL epitopes may thus be favoured for survival. Human cervical cancers are caused by papillomavirus infection, and these cancers consistently express the E7 protein of the oncogenic papillomavirus. We therefore investigated the MHC Class I restricted T cell epitopes of the human papillomavirus type 16 E7 oncoprotein using mice of five different genetic backgrounds, and an IFN-gamma ELISPOT assay, to determine the frequency with which MHC Class I epitopes might be expected in this small oncoprotein (98 amino acids). No MHC Class I restricted responses were detected in E7 immunized BALB/c (H-2d), CBA/CaH (H-2 k), FVB/N (H-2q) or A2KbH2b human HLA2.1 transgenic mice. In C57BL/6 J (H-2b) mice, a previously identified single antigenic epitope was detected. Therefore, we conclude that there is a paucity of MHC Class I restricted T cell epitopes in HPV16 E7 protein because of its small size. This might be advantageous to the virus. Furthermore here we present a quick and easy method to exhaustively determine CD8 T cell epitopes in proteins using a unique set of overlapping 8, 9 and 10 mer synthetic peptides.
Collapse
Affiliation(s)
- Vithagna Khammanivong
- Centre for Immunology and Cancer Research, University of Queensland, Princess Alexandra Hospital, Brisbane, Australia
| | | | | | | | | | | | | | | |
Collapse
|
364
|
|
365
|
Lin CW, Lee JY, Tsao YP, Shen CP, Lai HC, Chen SL. Oral vaccination with recombinant Listeria monocytogenes expressing human papillomavirus type 16 E7 can cause tumor growth in mice to regress. Int J Cancer 2002; 102:629-37. [PMID: 12448006 DOI: 10.1002/ijc.10759] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Listeria monocytogenes is a Gram-positive, facultative intracellular bacterium with the ability to present secreted proteins to the major histocompatibility complex class I pathway to stimulate cell-mediated immune response. In our study, we constructed the recombinant L. monocytogenes encoding human papillomavirus type 16 E7 gene (rLM-E7). When orally administered to syngeneic mice, rLM-E7 could induce a cytotoxic T-lymphocyte (CTL) response. Furthermore, in vitro flow cytometric assay and in vivo immune deficiency assays showed that rLM-E7 could prevent and eradicate tumor growth via CD8+-dependent CTLs. Hence, the potency of rLM-E7 as a therapeutic vaccine for cervical cancer is the result of the induction E7-specific cell-mediated immunity by L. monocytogenes. In addition to potency, this vaccine also offers ease of administration and reduced cost of production compared with other vaccines formulated for injection. Thus, L. monocytogenes encoding HPV-16 E7 may be a useful oral vaccine for cervical cancer treatment.
Collapse
Affiliation(s)
- Chih-Wei Lin
- Department of Microbiology & Immunology, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | | | | | | | | | | |
Collapse
|
366
|
van der Burg SH, de Jong A, Welters MJP, Offringa R, Melief CJM. The status of HPV16-specific T-cell reactivity in health and disease as a guide to HPV vaccine development. Virus Res 2002; 89:275-84. [PMID: 12445667 DOI: 10.1016/s0168-1702(02)00196-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Human papilloma viruses (HPV) are among the most common sexually transmitted pathogens in young adults. In the majority of individuals, anti-viral immunity is capable of suppressing viral infection but in a minority of patients viral infection is not cleared in time to prevent the development of malignancies. In these cases, HPV16-specific immunity may develop too late, is not strong enough, and/or is possibly of the wrong type. The influence of pre-existing immunity on the efficacy of vaccines is largely unknown. Nor has it been studied what the effect is of vaccines on the various types of pre-existing HPV-specific T-cell immunity. Animal models showing that vaccines are able to protect against a subsequent tumor challenge and even to treat transplantable tumors, are not qualified to address this point because tumor development is not preceded by persistent viral infection. Therefore, the comparison between fully characterized pre-existing HPV-specific immunity in patients and healthy subjects is a prerequisite for the full appreciation of vaccine-efficacy as well as for further development of next-generation vaccines.
Collapse
Affiliation(s)
- Sjoerd H van der Burg
- Tumor Immunology Group, Department of Immunohematology and Blood Transfusion, University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands.
| | | | | | | | | |
Collapse
|
367
|
Leggatt GR, Dunn LA, De Kluyver RL, Stewart T, Frazer IH. Interferon-gamma enhances cytotoxic T lymphocyte recognition of endogenous peptide in keratinocytes without lowering the requirement for surface peptide. Immunol Cell Biol 2002; 80:415-24. [PMID: 12225377 DOI: 10.1046/j.1440-1711.2002.01105.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Keratinocytes expressing the human papillomavirus (HPV) type 16 E7 protein, as a transgene driven by the K14 promoter, form a murine model of HPV-mediated epithelial cancers in humans. Our previous studies have shown that K14E7 transgenic skin grafts onto syngeneic mice are not susceptible to immune destruction despite the demonstrated presence of a strong, systemic CTL response directed against the E7 protein. Consistent with this finding, we now show that cultured, E7 transgenic keratinocytes (KC) express comparable endogenous levels of E7 protein to a range of CTL-sensitive E7-expressing cell lines but are not susceptible to CTL-mediated lysis in vitro. E7 transgenic and non-transgenic KC are susceptible to conventional mechanisms of CTL-mediated lysis, including perforin and Fas/FasL interaction when an excess of exogenous peptide is provided. The concentration of exogenous peptide required to render a cell susceptible to lysis was similar between KC and other conventional CTL targets (e.g. EL-4), despite large differences in H-2Db expression at the cell surface. Furthermore, exposure of KC to IFN-gamma increased H-2Db expression, but did not substantially alter the exogenous peptide concentration required to sensitize cells for half maximal lysis. In contrast, the lytic sensitivity of transgenic KC expressing endogenous E7 is modestly improved by exposure to IFN-gamma. Thus, failure of CTL to eliminate KC expressing endogenous E7, and by inference squamous tumours expressing E7, may reflect the need for a sustained, local inflammatory environment during the immune effector phase.
Collapse
MESH Headings
- Animals
- Antigen Presentation
- Carcinoma, Squamous Cell/immunology
- Carcinoma, Squamous Cell/virology
- Cells, Cultured/drug effects
- Cells, Cultured/immunology
- Cytotoxicity, Immunologic
- Disease Models, Animal
- Fas Ligand Protein
- H-2 Antigens/immunology
- Histocompatibility Antigen H-2D
- Interferon-gamma/pharmacology
- Keratinocytes/drug effects
- Keratinocytes/immunology
- Membrane Glycoproteins/deficiency
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/physiology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Oncogene Proteins, Viral/genetics
- Oncogene Proteins, Viral/immunology
- Papillomavirus E7 Proteins
- Peptide Fragments/immunology
- Perforin
- Pore Forming Cytotoxic Proteins
- Skin Neoplasms/immunology
- Skin Neoplasms/virology
- T-Lymphocytes, Cytotoxic/drug effects
- T-Lymphocytes, Cytotoxic/immunology
- fas Receptor/physiology
Collapse
Affiliation(s)
- Graham R Leggatt
- Centre for Immunology and Cancer Research, University of Queensland, Princess Alexandra Hospital, Brisbane, Australia.
| | | | | | | | | |
Collapse
|
368
|
Liu WJ, Gao F, Zhao KN, Zhao W, Fernando GJG, Thomas R, Frazer IH. Codon modified human papillomavirus type 16 E7 DNA vaccine enhances cytotoxic T-lymphocyte induction and anti-tumour activity. Virology 2002; 301:43-52. [PMID: 12359445 DOI: 10.1006/viro.2002.1584] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Polynucleotide immunisation with the E7 gene of human papillomavirus (HPV) type 16 induces only moderate levels of immune response, which may in part be due to limitation in E7 gene expression influenced by biased HPV codon usage. Here we compare for expression and immunogenicity polynucleotide expression plasmids encoding wild-type (pWE7) or synthetic codon optimised (pHE7) HPV16 E7 DNA. Cos-1 cells transfected with pHE7 expressed higher levels of E7 protein than similar cells transfected with pW7. C57BL/6 mice and F1 (C57x FVB) E7 transgenic mice immunised intradermally with E7 plasmids produced high levels of anti-E7 antibody. pHE7 induced a significantly stronger E7-specific cytotoxic T-lymphocyte response than pWE7 and 100% tumour protection in C57BL/6 mice, but neither vaccine induced CTL in partially E7 tolerant K14E7 transgenic mice. The data indicate that immunogenicity of an E7 polynucleotide vaccine can be enhanced by codon modification. However, this may be insufficient for priming E7 responses in animals with split tolerance to E7 as a consequence of expression of E7 in somatic cells.
Collapse
Affiliation(s)
- Wen Jun Liu
- Centre for Immunology and Cancer Research, University of Queensland, Princess Alexandra Hospital, Woolloongabba, Queensland 4102, Australia
| | | | | | | | | | | | | |
Collapse
|
369
|
Cheng WF, Hung CF, Pai SI, Hsu KF, He L, Ling M, Wu TC. Repeated DNA vaccinations elicited qualitatively different cytotoxic T lymphocytes and improved protective antitumor effects. J Biomed Sci 2002. [DOI: 10.1007/bf02254996] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
370
|
Gabrilovich DI, Cheng P, Fan Y, Yu B, Nikitina E, Sirotkin A, Shurin M, Oyama T, Adachi Y, Nadaf S, Carbone DP, Skoultchi AI. H1° histone and differentiation of dendritic cells. A molecular target for tumor‐derived factors. J Leukoc Biol 2002. [DOI: 10.1189/jlb.72.2.285] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
| | - Pingyan Cheng
- H. Lee Moffitt Cancer Center, University of South Florida, Tampa
| | - Yuhong Fan
- Department of Cell Biology and Cancer Center, Albert Einstein College of Medicine, Bronx, New York
| | - Bin Yu
- H. Lee Moffitt Cancer Center, University of South Florida, Tampa
| | | | - Allen Sirotkin
- Department of Cell Biology and Cancer Center, Albert Einstein College of Medicine, Bronx, New York
| | - Michael Shurin
- Department of Surgery, University of Pittsburgh, Pennsylvania; and
| | - Tsunehiro Oyama
- Department of Medicine and Cancer Center, Vanderbilt University, Nashville, Tennessee
| | - Yasushi Adachi
- Department of Medicine and Cancer Center, Vanderbilt University, Nashville, Tennessee
| | - Sorena Nadaf
- Department of Medicine and Cancer Center, Vanderbilt University, Nashville, Tennessee
| | - David P. Carbone
- Department of Medicine and Cancer Center, Vanderbilt University, Nashville, Tennessee
| | - Arthur I. Skoultchi
- Department of Cell Biology and Cancer Center, Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|
371
|
Street MD, Doan T, Herd KA, Tindle RW. Limitations of HLA-transgenic mice in presentation of HLA-restricted cytotoxic T-cell epitopes from endogenously processed human papillomavirus type 16 E7 protein. Immunology 2002; 106:526-36. [PMID: 12153516 PMCID: PMC1782761 DOI: 10.1046/j.1365-2567.2002.01442.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We investigated the use of mice transgenic for human leucocyte antigen (HLA) A*0201 antigen-binding domains to test vaccines composed of defined HLA A*0201-restricted cytotoxic T-lymphocyte (CTL) epitopes of human papillomavirus (HPV) type 16 E7 oncoprotein. HPV is detected in >90% of cervical carcinomas. HPV16 E7 oncoprotein transforms cells of the uterine cervix and functions as a tumour-associated antigen to which immunotherapeutic strategies may be directed. We report that although the HLA A*0201 E7 epitope peptides function both to prime for E7 CTL responses, and to sensitize target cells for E7-directed CTL killing in situations where antigen processing is not required, the epitopes are not processed out of either endogenously expressed or immunization-introduced E7, by the mouse antigen-processing and presentation machinery. Thus (1) CTL induced by HLA A*0201 peptide immunization killed E7 peptide-pulsed target cells, but did not kill target cells expressing whole E7; (2) immunization with whole E7 protein did not elicit CTL directed to HLA A*0201-restricted E7 CTL epitopes; (3) HLA A*0201-restricted CTL epitopes expressed in the context of a DNA polytope vaccine did not activate E7-specific T cells either in 'conventional' HLA A*0201 transgenic (A2.1Kb) mice, or in HHD transgenic mice in which expression of endogenous H-2 class 1 is precluded; and (4) HLA A*0201 E7 peptide epitope immunization was incapable of preventing the growth of an HLA A*0201- and E7-expressing tumour. There are generic implications for the universal applicability of HLA-class 1 transgenic mice for studies of human CTL epitope presentation in murine models of human infectious disease where recognition of endogenously processed antigen is necessary. There are also specific implications for the use of HLA A2 transgenic mice for the development of E7-based therapeutic vaccines for cervical cancer.
Collapse
Affiliation(s)
- Michael D Street
- Sir Albert Sakzewski Virus Research Centre, Royal Children's Hospital and Clinical Medical Virology Centre, University of Queensland, Brisbane, Australia
| | | | | | | |
Collapse
|
372
|
Zwaveling S, Ferreira Mota SC, Nouta J, Johnson M, Lipford GB, Offringa R, van der Burg SH, Melief CJM. Established human papillomavirus type 16-expressing tumors are effectively eradicated following vaccination with long peptides. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 169:350-8. [PMID: 12077264 DOI: 10.4049/jimmunol.169.1.350] [Citation(s) in RCA: 328] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Peptide-based vaccines aimed at the induction of effective T cell responses against established cancers have so far only met with limited clinical success and clearly need to be improved. In a preclinical model of human papillomavirus (HPV)16-induced cervical cancer we show that prime-boost vaccinations with the HPV16-derived 35 amino-acid long peptide E7(43-77), containing both a CTL epitope and a Th epitope, resulted in the induction of far more robust E7-specific CD8(+) T cell responses than vaccinations with the minimal CTL epitope only. We demonstrate that two distinct mechanisms are responsible for this effect. First, vaccinations with the long peptide lead to the generation of E7-specific CD4(+) Th cells. The level of the induced E7-specific CD8(+) T cell response proved to be dependent on the interactions of these Th cells with professional APC. Second, we demonstrate that vaccination with the long peptide and dendritic cell-activating agents resulted in a superior induction of E7-specific CD8(+) T cells, even when T cell help was excluded. This suggests that, due to its size, the long peptide was preferably endocytosed, processed, and presented by professional APCs. Moreover, the efficacy of this superior HPV-specific T cell induction was demonstrated in therapeutic prime-boost vaccinations in which the long peptide admixed with the dendritic cell-activating adjuvant oligodeoxynucleotide-CpG resulted in the eradication of large, established HPV16-expressing tumors. Because the vaccine types used in this study are easy to prepare under good manufacturing practice conditions and are safe to administer to humans, these data provide important information for future clinical trials.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Amino Acid Sequence
- Animals
- CD4-Positive T-Lymphocytes/immunology
- CD40 Antigens/metabolism
- CD40 Ligand/metabolism
- CD8-Positive T-Lymphocytes/immunology
- Cell Line, Transformed
- Cells, Cultured
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Disease Models, Animal
- Epitopes, T-Lymphocyte/immunology
- Histocompatibility Antigens Class II/immunology
- Humans
- Immunization, Secondary
- Injections, Subcutaneous
- Lymphocyte Activation/immunology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Molecular Sequence Data
- Oligodeoxyribonucleotides/administration & dosage
- Oligodeoxyribonucleotides/immunology
- Oncogene Proteins, Viral/administration & dosage
- Oncogene Proteins, Viral/biosynthesis
- Oncogene Proteins, Viral/immunology
- Papillomaviridae/immunology
- Papillomavirus E7 Proteins
- Papillomavirus Infections/immunology
- Papillomavirus Infections/prevention & control
- Peptide Fragments/administration & dosage
- Peptide Fragments/immunology
- Th1 Cells/immunology
- Tumor Cells, Cultured
- Tumor Virus Infections/immunology
- Tumor Virus Infections/prevention & control
- Vaccines, Subunit/administration & dosage
- Vaccines, Subunit/immunology
- Viral Vaccines/administration & dosage
- Viral Vaccines/immunology
Collapse
Affiliation(s)
- Sander Zwaveling
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
373
|
Micheletti F, Monini P, Fortini C, Rimessi P, Bazzaro M, Andreoni M, Giuliani M, Traniello S, Ensoli B, Gavioli R. Identification of cytotoxic T lymphocyte epitopes of human herpesvirus 8. Immunology 2002; 106:395-403. [PMID: 12100728 PMCID: PMC1782731 DOI: 10.1046/j.1365-2567.2002.01424.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The human herpesvirus 8 (HHV-8) is a human gamma2-herpesvirus that is implicated in the development of Kaposi's sarcoma (KS), primary effusion lymphoma and Castelman's disease. Since the responses of cytotoxic T lymphocytes (CTL) play a key role in the control of herpesvirus infection, it is important to identify and to characterize the CTL target epitopes of HHV-8 viral antigens. In this study, using peptide-binding motifs, we selected potential human leucocyte antigen (HLA)-A2-binding peptides from kaposin A and glycoprotein H (gH), that are latent and lytic HHV-8 antigens, respectively. HLA-A2-binding peptides were tested for their capacity to induce CTL responses in HHV-8-negative healthy donors. By this approach, we found that the majority of individuals responded to two HHV-8-derived CTL epitopes, namely, VLLNGWRWRL (amino acids 16-25), which derives from kaposin A, and FLNWQNLLNV (amino acids 59-68), which derives from gH. In addition, memory CTL responses to these epitopes were detected in disease-free individuals infected by HHV-8 demonstrating that the two epitopes are relevant targets of CTL-mediated immunity in vivo. The identified epitopes may be investigated for the development of immunotherapeutic strategies against HHV-8-associated malignancies.
Collapse
Affiliation(s)
- Fabiola Micheletti
- Department of Biochemistry and Molecular Biology, University of Ferrara, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
374
|
Parmiani G, Castelli C, Dalerba P, Mortarini R, Rivoltini L, Marincola FM, Anichini A. Cancer immunotherapy with peptide-based vaccines: what have we achieved? Where are we going? J Natl Cancer Inst 2002; 94:805-18. [PMID: 12048268 DOI: 10.1093/jnci/94.11.805] [Citation(s) in RCA: 282] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Many human tumor-associated antigens (TAAs) have recently been identified and molecularly characterized. When bound to major histocompatibility complex molecules, TAA peptides are recognized by T cells. Clinical studies have therefore been initiated to assess the therapeutic potential of active immunization or vaccination with TAA peptides in patients with metastatic cancer. So far, only a limited number of TAA peptides, mostly those recognized by CD8(+) T cells in melanoma patients, have been clinically tested. In some clinical trials, partial or complete tumor regression was observed in approximately 10%-30% of patients. No serious side effects have been reported. The clinical responses, however, were often not associated with a detectable T-cell-specific antitumor immune response when patients' T cells were evaluated in ex vivo assays. In this review, we analyze the available human TAA peptides, the potential immunogenicity (i.e., the ability to trigger a tumor-specific T-cell response) of TAA peptides in vitro and ex vivo, and the potential to construct slightly modified forms of TAA peptides that have increased T-cell stimulatory activity. We discuss the available data from clinical trials of TAA peptide-based vaccination (including those that used dendritic cells to present TAA peptides), identify possible reasons for the limited clinical efficacy of these vaccines, and suggest ways to improve the clinical outcome of TAA peptide-based vaccination for cancer patients.
Collapse
Affiliation(s)
- Giorgio Parmiani
- Unit of Immunotherapy of Human Tumors, Istituto Nazionale per lo Studio e la Cura dei Tumori, Milan, Italy.
| | | | | | | | | | | | | |
Collapse
|
375
|
Vandepapelière P. Therapeutic vaccination against chronic viral infections. THE LANCET. INFECTIOUS DISEASES 2002; 2:353-67. [PMID: 12144898 DOI: 10.1016/s1473-3099(02)00289-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Chronic viral infections such as those caused by hepatitis B virus, human papilloma virus, herpes simplex virus, and HIV, in theory, present logical targets of active specific immunotherapy. Indeed, immunological mechanisms are involved in several aspects of their pathogenesis and natural course, such as virus persistence, destruction of infected cells and control of viral replication. Therapeutic vaccination could therefore be an adequate replacement for, or adjunct to, existing therapies. Almost all approaches to therapeutic vaccination have been evaluated in those four disease areas. Despite encouraging results in animals none of these attempts has, so far, been completely successful in the human setting. However, with a better understanding of the immunological mechanisms involved in the control of disease successful therapeutic vaccines, used alone or in combination with other therapies, are an achievable goal.
Collapse
Affiliation(s)
- Pierre Vandepapelière
- Clinical R&D HIV vaccines and anti-infective therapeutic vaccines, GlaxoSmithKline Biologicals, Rue de l'Institut 89, B-1330, Rixensart, Belgium.
| |
Collapse
|
376
|
Steele JC, Roberts S, Rookes SM, Gallimore PH. Detection of CD4(+)- and CD8(+)-T-cell responses to human papillomavirus type 1 antigens expressed at various stages of the virus life cycle by using an enzyme-linked immunospot assay of gamma interferon release. J Virol 2002; 76:6027-36. [PMID: 12021335 PMCID: PMC136204 DOI: 10.1128/jvi.76.12.6027-6036.2002] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Human papillomavirus (HPV) antigens are expressed in epithelial cells at different stages of differentiation, and this may affect how they are handled by the immune system. We assessed the relative immunogenicities of four different HPV type 1 proteins: E6 and E7, which are made early in basal or parabasal cells; E4, which is made suprabasally in differentiating cells; and L1, a late protein which appears in the highly differentiated upper spinous layers. Pools of 15-mer peptides covering the primary sequences of all four proteins were used to screen 15 normal donors in enzyme-linked immunospot assays of gamma interferon release for both CD4(+)- and CD8(+)-T-cell reactivities. CD8(+)-T-cell responses were detected to the L1 protein in 7 of the 15 samples examined. No responses to E6, E7, or E4 were detected. CD4(+)-T-cell reactivities were again detected in 7 of the 15 donors. A broader spectrum of responses to E6 (three of seven), E4 (six of seven), and L1 (three of seven) was apparent, but there was no reactivity to E7. The predominant CD4(+) response was to E4. Reactivities were seen in some cases to corresponding regions on other common HPV types but were probably due to a multiple infection rather than to a cross-reaction. Antibodies to HPV1 virus-like particles were detected in 12 of the 15 (80%) donors, but antibody status did not correlate with T-cell reactivity. The differences in the relative immunogenicities of the four proteins revealed in this study are discussed in relation to how they may be processed and presented to the immune system by differentiating epithelial cells.
Collapse
Affiliation(s)
- Jane C Steele
- Cancer Research UK Institute for Cancer Studies, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom.
| | | | | | | |
Collapse
|
377
|
Wilcox RA, Flies DB, Zhu G, Johnson AJ, Tamada K, Chapoval AI, Strome SE, Pease LR, Chen L. Provision of antigen and CD137 signaling breaks immunological ignorance, promoting regression of poorly immunogenic tumors. J Clin Invest 2002. [PMID: 11877473 DOI: 10.1172/jci0214184] [Citation(s) in RCA: 175] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Treatment of advanced, poorly immunogenic tumors in animal models, considered the closest simulation available thus far for conditions observed in cancer patients, remains a major challenge for cancer immunotherapy. We reported previously that established tumors in mice receiving an agonistic mAb to the T cell costimulatory molecule 4-1BB (CD137) regress due to enhanced tumor antigen-specific cytotoxic T lymphocyte responses. In this study, we demonstrate that several poorly immunogenic tumors, including C3 tumor, TC-1 lung carcinoma, and B16-F10 melanoma, once established as solid tumors or metastases, are refractory to treatment by anti-4-1BB mAb. We provide evidence that immunological ignorance, rather than anergy or deletion, of tumor antigen--specific CTLs during the progressive growth of tumors prevents costimulation by anti-4-1BB mAb. Breaking CTL ignorance by immunization with a tumor antigen-derived peptide, although insufficient to stimulate a curative CTL response, is necessary for anti--4-1BB mAb to induce a CTL response leading to the regression of established tumors. Our results suggest a new approach for immunotherapy of human cancers.
Collapse
Affiliation(s)
- Ryan A Wilcox
- Department of Immunology, Mayo Clinic, Rochester, Minnesota 55905, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
378
|
Michel N, Osen W, Gissmann L, Schumacher TNM, Zentgraf H, Müller M. Enhanced immunogenicity of HPV 16 E7 fusion proteins in DNA vaccination. Virology 2002; 294:47-59. [PMID: 11886264 DOI: 10.1006/viro.2001.1321] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
DNA vaccination is a promising approach for inducing both humoral and cellular immune responses. For immunotherapy of HPV-16-associated diseases the E7 protein is considered a prime candidate, as it is expressed in all HPV-16-positive tumors. Unfortunately, the E7 protein is a very poor inducer of a cytotoxic T-cell response, when being used as antigen in DNA vaccination. Here we demonstrate that after fusion to protein export/import signals such as the herpes simplex virus ferry protein VP22, E7 can translocate in vitro from VP22-E7-expressing cells to neighboring cells that do not carry the VP22-E7 gene. In vivo, the VP22-E7 fusion shows significantly increased efficiency in inducing a cytotoxic T-cell response. Our data suggest that the export function of VP22 plays a major role in this phenomenon, since VP22 can be replaced by classical protein export signals, without impairing the induction of the E7-specific cellular immune response. However, all E7 fusion constructs showed significantly elevated protein steady-state levels, which might also account for the observed boost in immunogenicity.
Collapse
Affiliation(s)
- Nico Michel
- Institut für Angewandte Tumorvirologie, Deutsches Krebsforschungszentrum Heidelberg, Im Neuenheimer Feld 242, 69120 Heidelberg, Germany
| | | | | | | | | | | |
Collapse
|
379
|
Cheng WF, Hung CF, Hsu KF, Chai CY, He L, Polo JM, Slater LA, Ling M, Wu TC. Cancer immunotherapy using Sindbis virus replicon particles encoding a VP22-antigen fusion. Hum Gene Ther 2002; 13:553-68. [PMID: 11874633 DOI: 10.1089/10430340252809847] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Alphavirus vectors have emerged as a strategy for the development of cancer vaccines and gene therapy applications. The availability of a new packaging cell line (PCL), which is capable of generating alphavirus replicon particles without contamination from replication-competent virus, has advanced the field of vaccine development. This replication-defective vaccine vector has potential advantages over naked nucleic acid vaccines, such as increased efficiency of gene delivery and large-scale production. We have developed a new strategy to enhance nucleic acid vaccine potency by linking VP22, a herpes simplex virus type 1 (HSV-1) tegument protein, to a model antigen. This strategy facilitated the spread of linked E7 antigen to neighboring cells. In this study, we created a recombinant Sindbis virus (SIN)-based replicon particle encoding VP22 linked to a model tumor antigen, human papillomavirus type 16 (HPV-16) E7, using a stable SIN PCL. The linkage of VP22 to E7 in these SIN replicon particles resulted in a significant increase in the number of E7-specific CD8(+) T cell precursors and a strong antitumor effect against E7-expressing tumors in vaccinated C57BL/6 mice relative to wild-type E7 SIN replicon particles. Furthermore, a head-to-head comparison of VP22-E7-containing naked DNA, naked RNA replicons, or RNA replicon particle vaccines indicated that SINrep5-VP22/E7 replicon particles generated the most potent therapeutic antitumor effect. Our results indicated that the VP22 strategy used in the context of SIN replicon particles may facilitate the generation of a highly effective vaccine for widespread immunization.
Collapse
Affiliation(s)
- Wen-Fang Cheng
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD 21205, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
380
|
Wilcox RA, Flies DB, Zhu G, Johnson AJ, Tamada K, Chapoval AI, Strome SE, Pease LR, Chen L. Provision of antigen and CD137 signaling breaks immunological ignorance, promoting regression of poorly immunogenic tumors. J Clin Invest 2002; 109:651-9. [PMID: 11877473 PMCID: PMC150893 DOI: 10.1172/jci14184] [Citation(s) in RCA: 102] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Treatment of advanced, poorly immunogenic tumors in animal models, considered the closest simulation available thus far for conditions observed in cancer patients, remains a major challenge for cancer immunotherapy. We reported previously that established tumors in mice receiving an agonistic mAb to the T cell costimulatory molecule 4-1BB (CD137) regress due to enhanced tumor antigen-specific cytotoxic T lymphocyte responses. In this study, we demonstrate that several poorly immunogenic tumors, including C3 tumor, TC-1 lung carcinoma, and B16-F10 melanoma, once established as solid tumors or metastases, are refractory to treatment by anti-4-1BB mAb. We provide evidence that immunological ignorance, rather than anergy or deletion, of tumor antigen--specific CTLs during the progressive growth of tumors prevents costimulation by anti-4-1BB mAb. Breaking CTL ignorance by immunization with a tumor antigen-derived peptide, although insufficient to stimulate a curative CTL response, is necessary for anti--4-1BB mAb to induce a CTL response leading to the regression of established tumors. Our results suggest a new approach for immunotherapy of human cancers.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antigens, CD
- Antigens, Neoplasm/metabolism
- Female
- Humans
- Immune Tolerance
- Immunotherapy/methods
- Lymphocyte Activation/drug effects
- Mice
- Mice, Inbred C57BL
- Neoplasms, Experimental/immunology
- Neoplasms, Experimental/therapy
- Oncogene Proteins, Viral/immunology
- Papillomavirus E7 Proteins
- Receptors, Nerve Growth Factor/immunology
- Receptors, Tumor Necrosis Factor/immunology
- Signal Transduction/immunology
- T-Lymphocytes/drug effects
- T-Lymphocytes/immunology
- T-Lymphocytes, Cytotoxic/drug effects
- T-Lymphocytes, Cytotoxic/immunology
- Tumor Cells, Cultured
- Tumor Necrosis Factor Receptor Superfamily, Member 9
Collapse
Affiliation(s)
- Ryan A Wilcox
- Department of Immunology, Mayo Clinic, Rochester, Minnesota 55905, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
381
|
Hung CF, He L, Juang J, Lin TJ, Ling M, Wu TC. Improving DNA vaccine potency by linking Marek's disease virus type 1 VP22 to an antigen. J Virol 2002; 76:2676-82. [PMID: 11861834 PMCID: PMC135982 DOI: 10.1128/jvi.76.6.2676-2682.2002] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We have previously employed an intercellular spreading strategy using herpes simplex virus type 1 (HSV-1) VP22 protein to enhance DNA vaccine potency because DNA vaccines lack the intrinsic ability to amplify in cells. Recently, studies have demonstrated that the protein encoded by UL49 of Marek's disease virus type 1 (MDV-1) exhibits some degree of homology to the HSV-1 VP22 protein and features the property of intercellular transport. We therefore generated a DNA vaccine encoding MDV-1 VP22 linked to a model antigen, human papillomavirus type 16 E7. We demonstrated that compared with mice vaccinated with DNA encoding wild-type E7, mice vaccinated with MDV-1 VP22/E7 DNA exhibited a significant increase in number of gamma-interferon-secreting, E7-specific CD8(+)-T-cell precursors as well as stronger tumor prevention and treatment effects. Furthermore, our data indicated that the antitumor effect was CD8 dependent. These results suggested that the development of vaccines encoding VP22 fused to a target antigen might be a promising strategy for improving DNA vaccine potency.
Collapse
Affiliation(s)
- Chien-Fu Hung
- Pathology, The Johns Hopkins Medical Institutions, Baltimore, Maryland 21205, USA
| | | | | | | | | | | |
Collapse
|
382
|
Liu XS, Liu WJ, Zhao KN, Liu YH, Leggatt G, Frazer IH. Route of administration of chimeric BPV1 VLP determines the character of the induced immune responses. Immunol Cell Biol 2002; 80:21-9. [PMID: 11869359 DOI: 10.1046/j.1440-1711.2002.01051.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
To examine the mucosal immune response to papillomavirus virus-like particles (PV-VLP), mice were immunized with VLP intrarectally (i.r.), intravaginally (i.va.) or intramuscularly (i.m.) without adjuvant. PV-VLP were assembled with chimeric BPV-1 L1 proteins incorporating sequence from HIV-1 gp120, either the V3 loop or a shorter peptide incorporating a known CTL epitope (HIVP18I10). Antibody specific for BPV-1 VLP and P18 peptide was detected in serum following i.m., but not i.r. or i.va. immunization. Denatured VLP induced a much reduced immune response when compared with native VLP. Immune responses following mucosal administration of VLP were generally weaker than following systemic administration. VLP specific IgA was higher in intestine washes following i.r. than i.va. immunization, and higher in vaginal washes following i.m. than i.r. or i.va. immunization. No differences in specific antibody responses were seen between animals immunized with BPV-1 P18 VLP or with BPV-1 V3 VLP. Cytotoxic T lymphocyte precursors specific for the P18 CTL epitope were recovered from the spleen following i.m., i.va. or i.r. immunization with P18 VLP, and were similarly detected in Peyer's patches following i.m. or i.r. immunization. Thus, mucosal or systemic immunization with PV VLP induces mucosal CTL responses and this may be important for vaccines for mucosal infection with human papillomaviruses and for other viruses.
Collapse
Affiliation(s)
- Xiao Song Liu
- Centre for Immunology and Cancer Research,University of Queensland, Princess Alexandra Hospital, Woolloongabba, Queensland, Australia
| | | | | | | | | | | |
Collapse
|
383
|
Jiang S, Borthwick NJ, Morrison P, Gao GF, Steward MW. Virus-specific CTL responses induced by an H-2K(d)-restricted, motif-negative 15-mer peptide from the fusion protein of respiratory syncytial virus. J Gen Virol 2002; 83:429-438. [PMID: 11807236 DOI: 10.1099/0022-1317-83-2-429] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
We describe 15-mer peptide P8:F92-106 from the F protein of respiratory syncytial virus (RSV) that can act as an MHC class I-restricted (H-2K(d)) epitope for RSV-specific CD8(+) CTL. This peptide is interesting because not only is it the first murine CTL epitope to be identified in the F protein but also because it does not contain a known allele-specific motif, as all 15 amino acids appear to be required for effective presentation to CTL. In in vitro MHC class I refolding experiments, peptide P8:F92-106 induced complex formation with H-2K(d) heavy chains and beta2-microglobulin. Immunization of BALB/c mice with P8:F92-106 resulted in the induction of peptide and RSV-specific CTL responses as well as peptide-specific proliferative responses. Following intranasal challenge with RSV, P8:F92-106-immunized mice showed a significant reduction in viral load in the lungs compared to that seen in unimmunized mice. Furthermore, passive transfer of purified CD8(+) lymphocytes into BALB/c scid mice prior to challenge with RSV also resulted in a reduction in the virus load in lungs of challenged mice. These results indicate the potential of synthetic peptide epitopes for the induction of protective immune responses against RSV infection.
Collapse
Affiliation(s)
- Shisong Jiang
- Immunology Unit, Department of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK1
| | - Nicola J Borthwick
- Immunology Unit, Department of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK1
| | - Paul Morrison
- Immunology Unit, Department of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK1
| | - George F Gao
- Laboratory of Molecular Medicine, Children's Hospital, Boston MA 02115, USA2
| | - Michael W Steward
- Immunology Unit, Department of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK1
| |
Collapse
|
384
|
Vaccine Strategies for Colorectal Cancer. COLORECTAL CANCER 2002. [DOI: 10.1007/978-1-59259-160-2_41] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
385
|
Huarte E, Sarobe P, Lasarte JJ, Brem G, Weiss EH, Prieto J, Borrás-Cuesta F. Identification of HLA-B27-restricted cytotoxic T lymphocyte epitope from carcinoembryonic antigen. Int J Cancer 2002; 97:58-63. [PMID: 11774244 DOI: 10.1002/ijc.1579] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Characterization of epitopes recognized by cytotoxic T lymphocytes (CTLs) in the sequence of tumor antigens is an important step in the development of tumor therapies. Because carcinoembryonic antigen (CEA) is a protein expressed in a high number of epithelial tumors, it is an interesting target to study. We screened for the presence of HLA-B27-restricted CTL epitopes from CEA by studying the binding to HLA-B27 of 31 synthetic peptides predicted to bind to this molecule. This afforded 16 peptides with moderate or high binding affinity. Immunization of HLA-B27 transgenic mice with the best binder peptides yielded 4 immunogenic peptides: CEA(9-17), CEA(9-18), CEA(138-146) and CEA(360-369). However, splenocytes from mice immunized with a vaccinia virus-expressing CEA recognized only CEA(9-18). These CTLs were of the CD8(+) phenotype, which upon stimulation with peptide specifically produced IFN-gamma. Moreover, they did not cross-react against peptides of region 9-18 from proteins of the CEA family. Our results show that CEA(9-18) may induce specific CTL responses against CEA.
Collapse
Affiliation(s)
- Eduardo Huarte
- Departamento de Medicina Interna, Universidad de Navarra, Pamplona, Spain
| | | | | | | | | | | | | |
Collapse
|
386
|
Nikitina EY, Gabrilovich DI. Combination of gamma-irradiation and dendritic cell administration induces a potent antitumor response in tumor-bearing mice: approach to treatment of advanced stage cancer. Int J Cancer 2001; 94:825-33. [PMID: 11745485 DOI: 10.1002/1097-0215(20011215)94:6<825::aid-ijc1545>3.0.co;2-5] [Citation(s) in RCA: 112] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
We investigated a new approach to the treatment of advanced stage cancer, a combination of apoptosis-inducing therapy and dendritic cell (DC) administration. MethA sarcoma and C3 tumor containing defined tumor-specific antigens in form of peptides' epitopes were selected as experimental mouse models. Sites of established subcutaneous tumors were gamma-irradiated with 10 Gy 3-5 times with 4-5 day interval. DCs generated from bone marrow of syngeneic mice were injected i.v. and s.c. after each irradiation. A large number of cell tracker-labeled DCs accumulated at the site of the irradiated tumor after s.c. injections. This effect was not observed in non-irradiated tumors. Almost all of these DCs bound GFP-labeled MethA cells in tumor tissue. Interferon-gamma production by splenocytes in response to the tumor-specific MHC class I matched peptides was determined by ELISPOT assays. The combination of gamma-irradiation and DC administration, but not each of the treatments alone resulted in a significant increase in T cell response to the specific, but not to the control peptides. An increased proportion of tumor peptide-specific CD8(+)-cells was found only in that group of mice using staining with tetramers. DCs with defective presentation of antigen via MHC class I or MHC class II pathways were unable to induce peptide-specific T cell response. The combination of gamma-irradiation and DC administration but not each of the treatments alone resulted in a dramatic antitumor effect. A substantial proportion of mice completely rejected their tumors (80% in case of MethA sarcoma and 40% in case of C3 tumor). In the rest of the mice, tumor growth was notably suppressed or completely blocked. These data suggest that the combination of apoptosis-inducing therapy and DC administration may be an attractive approach to the treatment of advanced cancer.
Collapse
Affiliation(s)
- E Y Nikitina
- H. Lee Moffitt Cancer Center, University of South Florida, Tampa, FL 33612, USA
| | | |
Collapse
|
387
|
Nehete PN, Chitta S, Hossain MM, Hill L, Bernacky BJ, Baze W, Arlinghaus RB, Sastry KJ. Protection against chronic infection and AIDS by an HIV envelope peptide-cocktail vaccine in a pathogenic SHIV-rhesus model. Vaccine 2001; 20:813-25. [PMID: 11738745 DOI: 10.1016/s0264-410x(01)00408-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Based on our prior studies in mouse, monkey, chimpanzee, and human experimental systems, we identified six peptides encoded by highly conserved regions of the human immunodeficiency virus type 1 (HIV-1) envelope gene that selectively induce cellular immune responses in the absence of anti-viral antibody production. We tested a cocktail of the six peptides as a prototype vaccine for protection from simian human immunodeficiency virus (SHIV) infection and acquired immunodeficiency syndrome (AIDS) in a rhesus monkey model. Three monkeys were vaccinated with the peptide cocktail in Freund's adjuvant followed by autologous dendritic cells (DC) pulsed with these peptides. All the vaccinated animals exhibited significant induction of T-cell proliferation and cytotoxic T lymphocytes (CTL) responses, but no neutralizing antibodies. Two control mock-vaccinated monkeys showed no specific immune responses. Upon challenge with the pathogenic SHIV(KU-2), both the control and vaccinated monkeys were infected, but efficient clearance of virus-infected cells was observed in all the three vaccinated animals within 14 weeks. These animals also experienced a boosting of antiviral cellular immune responses after infection, and maintained antigen-specific IFN-gamma-producing cells in circulation beyond 42 weeks post-challenge. In contrast, the two mock-vaccinated monkeys had low to undetectable cellular immune responses and maintained significant levels of viral-infected cells and infectious virus in circulation. Further, in both the control monkeys plasma viremia was detectable beyond 38 weeks post-challenge indicating chronic phase infection. In one control monkey, the CD4+ cells dropped to very low levels by 2 weeks post-challenge and became undetectable by week 39 coinciding with high plasma viremia and AIDS, which included cachexia and ataxia. These results serve as proof of principle for the effectiveness of the HIV envelope peptide cocktail vaccine against chronic infection and AIDS, and support the development of multivalent peptide-based vaccine as a viable strategy to induce cell-mediated immunity (CMI) for protection against HIV and AIDS in humans.
Collapse
Affiliation(s)
- P N Nehete
- Department of Veterinary Sciences, The University of Texas M.D. Anderson Cancer Center, Science Park, 650 Coolwater Drive, Bastrop, TX 78602, USA
| | | | | | | | | | | | | | | |
Collapse
|
388
|
Gunn GR, Zubair A, Peters C, Pan ZK, Wu TC, Paterson Y. Two Listeria monocytogenes vaccine vectors that express different molecular forms of human papilloma virus-16 (HPV-16) E7 induce qualitatively different T cell immunity that correlates with their ability to induce regression of established tumors immortalized by HPV-16. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 167:6471-9. [PMID: 11714814 DOI: 10.4049/jimmunol.167.11.6471] [Citation(s) in RCA: 233] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Two recombinant Listeria monocytogenes (rLm) strains were produced that secrete the human papilloma virus-16 (HPV-16) E7 protein expressed in HPV-16-associated cervical cancer cells. One, Lm-E7, expresses and secretes E7 protein, whereas a second, Lm-LLO-E7, secretes E7 as a fusion protein joined to a nonhemolytic listeriolysin O (LLO). Lm-LLO-E7, but not Lm-E7, induces the regression of the E7-expressing tumor, TC-1, established in syngeneic C57BL/6 mice. Both recombinant E7-expressing rLm vaccines induce measurable anti-E7 CTL responses that stain positively for H-2D(b) E7 tetramers. Depletion of the CD8+ T cell subset before treatment abrogates the ability of Lm-LLO-E7 to impact on tumor growth. In addition, the rLm strains induce markedly different CD4+ T cell subsets. Depletion of the CD4+ T cell subset considerably reduces the ability of Lm-LLO-E7 to eliminate established TC-1 tumors. Surprisingly, the reverse is the case for Lm-E7, which becomes an effective anti-tumor immunotherapeutic in mice lacking this T cell subset. Ab-mediated depletion of TGF-beta and CD25+ cells improves the effectiveness of Lm-E7 treatment, suggesting that TGF-beta and CD25+ cells are in part responsible for this suppressive response. CD4+ T cells from mice immunized with Lm-E7 are capable of suppressing the ability of Lm-LLO-E7 to induce the regression of TC-1 when transferred to tumor-bearing mice. These studies demonstrate the complexity of L. monocytogenes-mediated tumor immunotherapy targeting the human tumor Ag, HPV-16 E7.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/immunology
- Bacterial Toxins
- Bacterial Vaccines/genetics
- Bacterial Vaccines/immunology
- CD4-Positive T-Lymphocytes/transplantation
- CD8-Positive T-Lymphocytes/immunology
- Cancer Vaccines/genetics
- Cancer Vaccines/immunology
- Cell Line, Transformed/immunology
- Cell Line, Transformed/pathology
- Cell Line, Transformed/virology
- Cytotoxicity, Immunologic/genetics
- Epitopes, T-Lymphocyte/immunology
- Gene Expression Regulation, Viral/immunology
- Genetic Vectors/chemical synthesis
- Genetic Vectors/immunology
- Heat-Shock Proteins/genetics
- Heat-Shock Proteins/immunology
- Hemolysin Proteins
- Humans
- Interferon-gamma/physiology
- Listeria monocytogenes/genetics
- Listeria monocytogenes/immunology
- Listeria monocytogenes/metabolism
- Lymphocyte Activation/genetics
- Mice
- Mice, Inbred C57BL
- Oncogene Proteins, Viral/biosynthesis
- Oncogene Proteins, Viral/genetics
- Oncogene Proteins, Viral/immunology
- Oncogene Proteins, Viral/metabolism
- Papillomaviridae/genetics
- Papillomaviridae/immunology
- Papillomavirus E7 Proteins
- Papillomavirus Vaccines
- Spleen/cytology
- Spleen/immunology
- T-Lymphocytes, Cytotoxic/immunology
- Transforming Growth Factor beta/immunology
- Transforming Growth Factor beta/metabolism
- Tumor Cells, Cultured/immunology
- Tumor Cells, Cultured/pathology
- Tumor Cells, Cultured/virology
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
Collapse
Affiliation(s)
- G R Gunn
- Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | | | | | | | | | | |
Collapse
|
389
|
Carrasco D, Straten MV, Tyring SK. Prophylactic and therapeutic vaccines for genital papillomavirus infection. J Investig Dermatol Symp Proc 2001; 6:238-43. [PMID: 11924835 DOI: 10.1046/j.0022-202x.2001.00036.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 development of potential therapeutic and prophylactic vaccines for human papillomavirus (HPV) infection is a very exciting area of HPV research. There are a number of features of HPV biology that makes the development of a vaccine particularly difficult, although there are several examples of vaccines that have had spectacular success in the prevention of other viral diseases. Our poor understanding of the immune response to HPV infection is the first problem. We do not understand the mechanism by which spontaneous clearing of warts is generated and therefore cannot particularly target this pathway in the development of a vaccine. Furthermore, there is no in vitro culture system nor an animal model for HPV. Another problem is that there is no ready source of live virus that might be exploited for a live attenuated viral vaccine, such as was used with poliovirus. Although most other viruses spend a portion of their life cycle in the systemic circulation where they are vulnerable to neutralizing antibodies, HPV remain exclusively in the epithelium and thus antibodies must transverse the basement membrane and reach the other layers of the skin or mucosa to be effective in preventing infection. Significant progress is being made in the development of potential vaccine candidates despite these and other confounding factors.
Collapse
Affiliation(s)
- D Carrasco
- Department of Dermatology, University of Texas Medical Branch, Galveston 77555-1070, USA
| | | | | |
Collapse
|
390
|
Frazer IH, De Kluyver R, Leggatt GR, Guo HY, Dunn L, White O, Harris C, Liem A, Lambert P. Tolerance or immunity to a tumor antigen expressed in somatic cells can be determined by systemic proinflammatory signals at the time of first antigen exposure. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 167:6180-7. [PMID: 11714778 DOI: 10.4049/jimmunol.167.11.6180] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Mice transgenic for the E7 tumor Ag of human papillomavirus type 16, driven from a keratin 14 promoter, express E7 in keratinocytes but not dendritic cells. Grafted E7-transgenic skin is not rejected by E7-immunized mice that reject E7-transduced transplantable tumors. Rejection of recently transplanted E7-transgenic skin grafts, but not of control nontransgenic grafts or of established E7-transgenic grafts, is induced by systemic administration of live or killed Listeria monocytogenes or of endotoxin. Graft recipients that reject an E7 graft reject a subsequent E7 graft more rapidly and without further L. monocytogenes exposure, whereas recipients of an E7 graft given without L. monocytogenes do not reject a second graft, even if given with L. monocytogenes. Thus, cross-presentation of E7 from keratinocytes to the adaptive immune system occurs with or without a proinflammatory stimulus, but proinflammatory stimuli at the time of first cross-presentation of Ag can determine the nature of the immune response to the Ag. Furthermore, immune effector mechanisms responsible for rejection of epithelium expressing a tumor Ag in keratinocytes are different from those that reject an E7-expressing transplantable tumor. These observations have implications for immunotherapy for epithelial cancers.
Collapse
MESH Headings
- Animals
- Antigen Presentation/genetics
- Antigens, Bacterial/administration & dosage
- Antigens, Bacterial/immunology
- Antigens, Neoplasm/biosynthesis
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/metabolism
- Cells, Cultured
- Female
- Graft Rejection/genetics
- Graft Rejection/immunology
- Immune Tolerance/genetics
- Inflammation/genetics
- Inflammation/immunology
- Injections, Intravenous
- Keratinocytes/immunology
- Keratinocytes/metabolism
- Listeriosis/genetics
- Listeriosis/immunology
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Oncogene Proteins, Viral/biosynthesis
- Oncogene Proteins, Viral/genetics
- Oncogene Proteins, Viral/immunology
- Papillomaviridae/genetics
- Papillomaviridae/immunology
- Papillomavirus E7 Proteins
- Signal Transduction/genetics
- Signal Transduction/immunology
- Skin Transplantation/immunology
- Skin Transplantation/methods
- Time Factors
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- I H Frazer
- Centre for Immunology and Cancer Research, University of Queensland, Brisbane, Australia.
| | | | | | | | | | | | | | | | | |
Collapse
|
391
|
Charo J, Geluk A, Sundbäck M, Mirzai B, Diehl AD, Malmberg KJ, Achour A, Huriguchi S, van Meijgaarden KE, Drijfhout JW, Beekman N, van Veelen P, Ossendorp F, Ottenhoff TH, Kiessling R. The identification of a common pathogen-specific HLA class I A*0201-restricted cytotoxic T cell epitope encoded within the heat shock protein 65. Eur J Immunol 2001; 31:3602-11. [PMID: 11745380 DOI: 10.1002/1521-4141(200112)31:12<3602::aid-immu3602>3.0.co;2-l] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Bacterial antigens recognized by CD8(+) T cells in the context of MHC class I are thought to play a crucial role in protection against pathogenic intracellular bacteria. Here, we demonstrate the induction of HLA-A*0201-restricted CD8(+) T cell responses against six new high-affinity HLA-A*0201-binding CTL epitopes, encoded within an immunodominant and highly conserved antigen of Mycobacteria, the heat shock protein 65 (hsp65). One of these epitopes, Mhsp65(9(369)), is identical in a large number of pathogenic bacteria, and is recognized in a CD8-independent fashion. Mhsp65(9(369)) could be presented by either mycobacterial hsp65-pulsed target cells or BCG-infected macrophages. Interestingly, T cells specific for this epitope did not recognize the corresponding human hsp65 homologue, probably due to structural differences as revealed by modeling studies. Furthermore, in vitro proteasome digestion analyses show that, whereas the mycobacterial hsp65 epitope is efficiently generated, the human hsp65 homologue is not, thus avoiding the induction of autoreactivity. Collectively, these findings describe high-affinity HLA class I-binding epitopes that are naturally processed and are recognized efficiently by MHC class I-restricted CD8(+) T cells, providing a rational basis for the development of subunit vaccine strategies against tuberculosis and other intracellular infectious diseases.
Collapse
Affiliation(s)
- J Charo
- Microbiology and Tumorbiology Center, Karolinska Institute, Stockholm, Sweden.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
392
|
Kanduc D, Lucchese A, Mittelman A. Individuation of monoclonal anti-HPV16 E7 antibody linear peptide epitope by computational biology. Peptides 2001; 22:1981-5. [PMID: 11786180 DOI: 10.1016/s0196-9781(01)00539-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We applied computational biology to identify the linear amino acid sequence recognized by a mouse monoclonal antibody raised against the full length HPV16 E7 oncoprotein. Computer-assisted search for the epitopic peptide used two parameters: the capability of E7 peptides to bind to MHC class II molecules, and the similarity level of the oncoprotein sequence to the mouse proteome. We report that anti-E7 mAb recognized the peptide having both high binding potential to MHC II molecules and low level of molecular mimicry to mouse proteome. Peptide ability to bind to MHC II molecules appears a necessary but not sufficient condition to determine peptide immunodominance, by needing to be supported by a low degree of peptide similarity to the host's proteome.
Collapse
Affiliation(s)
- D Kanduc
- CARSO Cancer Research Center, University of Bari, Italy.
| | | | | |
Collapse
|
393
|
Lapteva N, Nieda M, Ando Y, Nicol A, Ide K, Yamaura A, Hatta-Ohashi Y, Egawa K, Juji T, Tokunaga K. Gene expression analysis in human monocytes, monocyte-derived dendritic cells, and alpha-galactosylceramide-pulsed monocyte-derived dendritic cells. Biochem Biophys Res Commun 2001; 289:531-8. [PMID: 11716506 DOI: 10.1006/bbrc.2001.6003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
In vitro proliferation and functional activation of V alpha 24NKT cells following stimulation with alpha-galactosylceramide (alpha-GalCer)-pulsed dendritic cells (DCs) have been observed. Because little is known about the molecular events on DCs following interaction with alpha-GalCer, we performed gene expression profiling of 2400 genes in monocytes and monocyte-derived immature DCs pulsed with alpha-GalCer (alpha-GalCer-imDCs). Overall, the expression levels of 48 genes were up-regulated and 28 were down-regulated in alpha-GalCer-imDCs. Semiquantitative RT-PCR analysis on monocytes, imDCs, alpha-GalCer-imDCs, and mature DCs confirmed the up- and down-regulation of the mRNA expression levels of 28 selected genes. Notably, we identified the specific up-regulation of mRNA expression levels of ribonuclease A and collapsin response mediator protein upon the stimulation of imDC with alpha-GalCer, suggesting a novel immunomodulating effect of alpha-GalCer on imDCs. In this study, we used imDCs prepared by culturing of monocytes with GM-CSF and IL-4 for 5 days and mDCs prepared by further culturing of imDCs with TNF alpha for two extra days.
Collapse
Affiliation(s)
- N Lapteva
- Department of Human Genetics, University of Tokyo, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
394
|
Da Silva DM, Pastrana DV, Schiller JT, Kast WM. Effect of preexisting neutralizing antibodies on the anti-tumor immune response induced by chimeric human papillomavirus virus-like particle vaccines. Virology 2001; 290:350-60. [PMID: 11883199 DOI: 10.1006/viro.2001.1179] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Chimeric human papillomavirus virus-like particles (HPV cVLPs) carrying HPV16 E7 protein are potent vaccines for inducing cell-mediated immunity (CMI) against HPV-induced tumors in animal models. We tested the hypothesis that virion-neutralizing antibodies generated during an initial vaccination might prevent effective boosting of CMI to the cVLPs. Mice with circulating HPV16-neutralizing antibodies, generated by direct immunization with wild-type VLPs or by passive transfer of hyperimmune anti-HPV16 VLP mouse sera, were subsequently vaccinated with HPV16 E7-containing cVLPs. Mice with preexisting neutralizing antibodies were not protected from HPV16 E7-positive TC-1 tumor challenge, compared to the protection seen in mice lacking these antibodies. Antibody-coated VLPs bound very inefficiently to receptor-positive cell lines, suggesting that one of the mechanisms of antibody interference is blocking of VLP binding to its receptor and thereby uptake of VLPs by antigen-presenting cells. Our results suggest that repetitive vaccination with a cVLP for induction of cellular immune responses to an incorporated antigen may be of limited effectiveness due to the presence of neutralizing antibodies against the capsid proteins induced after the first application. This limitation could potentially be overcome by boosting with cVLPs containing the same target antigen incorporated into other papillomavirus-type VLPs.
Collapse
Affiliation(s)
- D M Da Silva
- Cancer Immunology Program, Cardinal Bernardin Cancer Center, Department of Microbiology and Immunology, Loyola University Chicago, 2160 South First Avenue, Maywood, Illinois 60153, USA
| | | | | | | |
Collapse
|
395
|
Abstract
Cancer is one of the leading causes of death in Western society. Despite improvements in screening, diagnosis and treatment of cancer, many patients ultimately succumb to their disease. Advances in molecular biology and our increased understanding of how the immune system functions have led to an intense interest in the development of cancer vaccines.
Collapse
Affiliation(s)
- A C Armstrong
- CRC Department of Medical Oncology, University of Manchester & Paterson Institute for Cancer Research, Christie Hospital NHS Trust, Manchester M20 4BX, UK
| | | |
Collapse
|
396
|
Lamikanra A, Pan ZK, Isaacs SN, Wu TC, Paterson Y. Regression of established human papillomavirus type 16 (HPV-16) immortalized tumors in vivo by vaccinia viruses expressing different forms of HPV-16 E7 correlates with enhanced CD8(+) T-cell responses that home to the tumor site. J Virol 2001; 75:9654-64. [PMID: 11559797 PMCID: PMC114536 DOI: 10.1128/jvi.75.20.9654-9664.2001] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2000] [Accepted: 06/20/2001] [Indexed: 02/02/2023] Open
Abstract
Using vaccinia virus as a live vector, we show that the expression of human papillomavirus type 16 (HPV-16) E7 fused to a nonhemolytic portion of the Listeria monocytogenes virulence factor, listeriolysin O (LLO), induces an immune response that causes the regression of established HPV-16 immortalized tumors in C57BL/6 mice. The vaccinia virus construct expressing LLO fused to E7 (VacLLOE7) was compared with two previously described vaccinia virus constructs: one that expresses unmodified E7 (VacE7) and another that expresses E7 in a form designed to direct it to intracellular lysosomal compartments and improve major histocompatibility complex class II-restricted responses (VacSigE7LAMP-1). C57BL/6 mice bearing established HPV-16 immortalized tumors of 5 or 8 mm were treated with each of these vaccines. Fifty percent of the mice treated with VacLLOE7 remained tumor free 2 months after tumor inoculation, whereas 12 to 25% of the mice were tumor free after treatment with VacSigE7LAMP-1 (depending on the size of the tumor). No mice were tumor free in the group given VacE7. Compared to VacE7, VacSigE7LAMP-1 and VacLLOE7 resulted in increased numbers of H2-D(b)-specific tetramer-positive CD8(+) T cells in mouse spleens that produced gamma interferon and tumor necrosis factor alpha upon stimulation with RAHYNIVTF peptide. In addition, the highest frequency of tetramer-positive T cells was seen in the tumor sites of mice treated with VacLLOE7. An increased efficiency of E7-specific lysis by splenocytes from mice immunized with VacLLOE7 was also observed. These results indicate that the fusion of E7 with LLO not only enhances antitumor therapy by improving the tumoricidal function of E7-specific CD8(+) T cells but may also increase the number of antigen-specific CD8(+) T cells in the tumor, the principle site of antigen expression.
Collapse
Affiliation(s)
- A Lamikanra
- Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | |
Collapse
|
397
|
Cheng WF, Hung CF, Chai CY, Hsu KF, He L, Ling M, Wu TC. Tumor-specific immunity and antiangiogenesis generated by a DNA vaccine encoding calreticulin linked to a tumor antigen. J Clin Invest 2001. [PMID: 11544272 DOI: 10.1172/jci200112346] [Citation(s) in RCA: 217] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Antigen-specific cancer immunotherapy and antiangiogenesis have emerged as two attractive strategies for cancer treatment. An innovative approach that combines both mechanisms will likely generate the most potent antitumor effect. We tested this approach using calreticulin (CRT), which has demonstrated the ability to enhance MHC class I presentation and exhibit an antiangiogenic effect. We explored the linkage of CRT to a model tumor antigen, human papilloma virus type-16 (HPV-16) E7, for the development of a DNA vaccine. We found that C57BL/6 mice vaccinated intradermally with CRT/E7 DNA exhibited a dramatic increase in E7-specific CD8(+) T cell precursors and an impressive antitumor effect against E7-expressing tumors compared with mice vaccinated with wild-type E7 DNA or CRT DNA. Vaccination of CD4/CD8 double-depleted C57BL/6 mice and immunocompromised (BALB/c nu/nu) mice with CRT/E7 DNA or CRT DNA generated significant reduction of lung tumor nodules compared with wild-type E7 DNA, suggesting that antiangiogenesis may have contributed to the antitumor effect. Examination of microvessel density in lung tumor nodules and an in vivo angiogenesis assay further confirmed the antiangiogenic effect generated by CRT/E7 and CRT. Thus, cancer therapy using CRT linked to a tumor antigen holds promise for treating tumors by combining antigen-specific immunotherapy and antiangiogenesis.
Collapse
Affiliation(s)
- W F Cheng
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, Maryland 21205, USA
| | | | | | | | | | | | | |
Collapse
|
398
|
Cheng WF, Hung CF, Chai CY, Hsu KF, He L, Ling M, Wu TC. Tumor-specific immunity and antiangiogenesis generated by a DNA vaccine encoding calreticulin linked to a tumor antigen. J Clin Invest 2001; 108:669-78. [PMID: 11544272 PMCID: PMC209378 DOI: 10.1172/jci12346] [Citation(s) in RCA: 107] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Antigen-specific cancer immunotherapy and antiangiogenesis have emerged as two attractive strategies for cancer treatment. An innovative approach that combines both mechanisms will likely generate the most potent antitumor effect. We tested this approach using calreticulin (CRT), which has demonstrated the ability to enhance MHC class I presentation and exhibit an antiangiogenic effect. We explored the linkage of CRT to a model tumor antigen, human papilloma virus type-16 (HPV-16) E7, for the development of a DNA vaccine. We found that C57BL/6 mice vaccinated intradermally with CRT/E7 DNA exhibited a dramatic increase in E7-specific CD8(+) T cell precursors and an impressive antitumor effect against E7-expressing tumors compared with mice vaccinated with wild-type E7 DNA or CRT DNA. Vaccination of CD4/CD8 double-depleted C57BL/6 mice and immunocompromised (BALB/c nu/nu) mice with CRT/E7 DNA or CRT DNA generated significant reduction of lung tumor nodules compared with wild-type E7 DNA, suggesting that antiangiogenesis may have contributed to the antitumor effect. Examination of microvessel density in lung tumor nodules and an in vivo angiogenesis assay further confirmed the antiangiogenic effect generated by CRT/E7 and CRT. Thus, cancer therapy using CRT linked to a tumor antigen holds promise for treating tumors by combining antigen-specific immunotherapy and antiangiogenesis.
Collapse
MESH Headings
- Animals
- Antibodies, Neoplasm/biosynthesis
- Antigen Presentation
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/metabolism
- Calcium-Binding Proteins/genetics
- Calcium-Binding Proteins/metabolism
- Calreticulin
- Cancer Vaccines
- Cells, Cultured
- DNA, Neoplasm/genetics
- Endoplasmic Reticulum/metabolism
- Lung Neoplasms/pathology
- Lung Neoplasms/secondary
- Lymphocyte Depletion
- Mice
- Mice, Inbred C57BL
- Mice, Nude
- Neoplasms/immunology
- Neoplasms/pathology
- Neoplasms/therapy
- Neovascularization, Pathologic
- Oncogene Proteins, Viral/genetics
- Oncogene Proteins, Viral/metabolism
- Papillomavirus E7 Proteins
- Ribonucleoproteins/genetics
- Ribonucleoproteins/metabolism
- T-Lymphocytes, Cytotoxic/immunology
- Tumor Cells, Cultured
- Vaccines, DNA
Collapse
Affiliation(s)
- W F Cheng
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, Maryland 21205, USA
| | | | | | | | | | | | | |
Collapse
|
399
|
den Boer AT, Diehl L, van Mierlo GJ, van der Voort EI, Fransen MF, Krimpenfort P, Melief CJ, Offringa R, Toes RE. Longevity of antigen presentation and activation status of APC are decisive factors in the balance between CTL immunity versus tolerance. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 167:2522-8. [PMID: 11509591 DOI: 10.4049/jimmunol.167.5.2522] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Encounter of Ag by naive T cells can lead to T cell priming as well as tolerance. The balance between immunity and tolerance is controlled by the conditions of Ag encounter and the activation status of the APC. We have investigated the rules that govern this balance in case an environment that normally induces tolerance is reverted into a milieu that promotes T cell priming, using a minimal CTL epitope derived from human adenovirus type 5 E1A. Vaccination of mice s.c. with E1A peptide in IFA readily induces CTL tolerance, resulting in the inability to control E1A-expressing tumors. The present study shows that efficient CTL priming is achieved when this peptide vaccine is combined with systemic administration of APC-activating compounds like agonistic anti-CD40 mAb or polyriboinosinate-polyribocytidylate. Surprisingly, this CTL response is not long-lasting and therefore fails to protect against tumor outgrowth. Disappearance of CTL reactivity was strongly associated with systemic persistence of the peptide for >200 days. In contrast, peptide administered in PBS does not persist and generates long term CTL immunity capable of rejecting Ad5E1A-positive tumors, when combined with CD40 triggering. Thus, presentation of CTL epitopes in an appropriate costimulatory setting by activated APC, although being essential and sufficient for CTL priming, eventually results in tolerance when the Ag persists systemically for prolonged times. These observations are important for the development of immune intervention schemes in autoimmunity and cancer.
Collapse
MESH Headings
- Adenovirus E1A Proteins/administration & dosage
- Adenovirus E1A Proteins/immunology
- Animals
- Antigen Presentation
- Antigen-Presenting Cells/immunology
- CD40 Antigens/metabolism
- Humans
- Immune Tolerance
- Kinetics
- Lymphocyte Activation
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Neoplasms, Experimental/immunology
- Neoplasms, Experimental/therapy
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
- T-Lymphocytes, Cytotoxic/immunology
Collapse
Affiliation(s)
- A T den Boer
- Department of Immunohematology, Leiden University Medical Center, Leiden, The Netherlands.
| | | | | | | | | | | | | | | | | |
Collapse
|
400
|
Jäger D, Jäger E, Knuth A. Immune responses to tumour antigens: implications for antigen specific immunotherapy of cancer. J Clin Pathol 2001; 54:669-74. [PMID: 11533070 PMCID: PMC1731514 DOI: 10.1136/jcp.54.9.669] [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: 12/14/2022]
Abstract
Tumour associated antigens recognised by cellular or humoral effectors of the immune system are potential targets for antigen specific cancer immunotherapy. Different categories of cancer antigens have been identified that induce cytotoxic T lymphocyte (CTL) responses in vitro and in vivo, namely: (1) "cancer testis" (CT) antigens, expressed in different tumours and normal testis, (2) melanocyte differentiation antigens, (3) point mutations of normal genes, (4) self antigens that are overexpressed in malignant tissues, and (5) viral antigens. Clinical studies with peptides and proteins derived from these antigens have been initiated to study the efficacy of inducing specific CTL responses in vivo. Immunological and clinical parameters for the assessment of antigen specific immune responses have been defined-delayed type hypersensitivity (DTH), CTL, autoimmmune, and tumour regression responses. Specific DTH and CTL responses and tumour regression have been observed after the intradermal administration of tumour associated peptides alone. Peptide specific immune reactions were enhanced after using granulocyte macrophage stimulating factor (GM-CSF) as a systemic adjuvant by increasing the frequency of dermal antigen presenting Langerhans cells. Complete tumour regression has been observed in the context of measurable peptide specific CTL. However, in single cases with disease progression after an initial tumour response, either a loss of single antigens targeted by CTL or of the presenting major histocompatibility complex (MHC) class I allele was detected, pointing towards immunisation induced immune escape. Cytokines to modulate antigen and MHC class I expression in vivo are being evaluated to prevent immunoselection. Recently, a new CT antigen, NY-ESO-1, has been identified on the basis of spontaneous antibody responses to tumour associated antigens. NY-ESO-1 appears to be one of the most immunogenic antigens known to date, with spontaneous immune responses observed in 50% of patients with NY-ESO-1 expressing cancers. Clinical studies have been initiated to evaluate the immunogenicity of different NY-ESO-1 constructs to induce both humoral and cellular immune responses in vivo.
Collapse
Affiliation(s)
- D Jäger
- II. Medizinische Klinik, Hämatologie-Onkologie, Krankenhaus Nordwest, Steinbacher Hohl 2-26, 60488 Frankfurt am Main, Germany
| | | | | |
Collapse
|