1
|
Oelkrug C. Analysis of physical and biological delivery systems for DNA cancer vaccines and their translation to clinical development. Clin Exp Vaccine Res 2024; 13:73-82. [PMID: 38752006 PMCID: PMC11091436 DOI: 10.7774/cevr.2024.13.2.73] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 07/17/2023] [Accepted: 03/30/2024] [Indexed: 05/18/2024] Open
Abstract
DNA cancer vaccines as an approach in tumor immunotherapy are still being investigated in preclinical and clinical settings. Nevertheless, only a small number of clinical studies have been published so far and are still active. The investigated vaccines show a relatively stable expression in in-vitro transfected cells and may be favorable for developing an immunologic memory in patients. Therefore, DNA vaccines could be suitable as a prophylactic or therapeutic approach against cancer. Due to the low efficiency of these vaccines, the administration technique plays an important role in the vaccine design and its efficacy. These DNA cancer vaccine delivery systems include physical, biological, and non-biological techniques. Although the pre-clinical studies show promising results in the application of the different delivery systems, further studies in clinical trials have not yet been successfully proven.
Collapse
|
2
|
New synthetic quinaldine conjugates: Assessment of their anti-cholinesterase, anti-tyrosinase and cytotoxic activities, and molecular docking analysis. ARAB J CHEM 2022. [DOI: 10.1016/j.arabjc.2022.104177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
3
|
Modified Vaccinia Virus Ankara Can Induce Optimal CD8 + T Cell Responses to Directly Primed Antigens Depending on Vaccine Design. J Virol 2019; 93:JVI.01154-19. [PMID: 31375596 PMCID: PMC6803277 DOI: 10.1128/jvi.01154-19] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 07/29/2019] [Indexed: 01/19/2023] Open
Abstract
A variety of strains of vaccinia virus (VACV) have been used as recombinant vaccine vectors with the aim of inducing robust CD8+ T cell immunity. While much of the pioneering work was done with virulent strains, such as Western Reserve (WR), attenuated strains such as modified vaccinia virus Ankara (MVA) are more realistic vectors for clinical use. To unify this literature, side-by-side comparisons of virus strains are required. Here, we compare the form of antigen that supports optimal CD8+ T cell responses for VACV strains WR and MVA using equivalent constructs. We found that for multiple antigens, minimal antigenic constructs (epitope minigenes) that prime CD8+ T cells via the direct presentation pathway elicited optimal responses from both vectors, which was surprising because this finding contradicts the prevailing view in the literature for MVA. We then went on to explore the discrepancy between current and published data for MVA, finding evidence that the expression locus and in some cases the presence of the viral thymidine kinase may influence the ability of this strain to prime optimal responses from antigens that require direct presentation. This extends our knowledge of the design parameters for VACV vectored vaccines, especially those based on MVA.IMPORTANCE Recombinant vaccines based on vaccinia virus and particularly attenuated strains such as MVA are in human clinical trials, but due to the complexity of these large vectors much remains to be understood about the design parameters that alter their immunogenicity. Previous work had found that MVA vectors should be designed to express stable protein in order to induce robust immunity by CD8+ (cytotoxic) T cells. Here, we found that the primacy of stable antigen is not generalizable to all designs of MVA and may depend where a foreign antigen is inserted into the MVA genome. This unexpected finding suggests that there is an interaction between genome location and the best form of antigen for optimal T cell priming in MVA and thus possibly other vaccine vectors. It also highlights that our understanding of antigen presentation by even the best studied of vaccine vectors remains incomplete.
Collapse
|
4
|
MacNeill AL, Weishaar KM, Séguin B, Powers BE. Safety of an Oncolytic Myxoma Virus in Dogs with Soft Tissue Sarcoma. Viruses 2018; 10:v10080398. [PMID: 30060548 PMCID: PMC6115854 DOI: 10.3390/v10080398] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 07/25/2018] [Accepted: 07/27/2018] [Indexed: 12/21/2022] Open
Abstract
Many oncolytic viruses that are efficacious in murine cancer models are ineffective in humans. The outcomes of oncolytic virus treatment in dogs with spontaneous tumors may better predict human cancer response and improve treatment options for dogs with cancer. The objectives of this study were to evaluate the safety of treatment with myxoma virus lacking the serp2 gene (MYXVΔserp2) and determine its immunogenicity in dogs. To achieve these objectives, dogs with spontaneous soft tissue sarcomas were treated with MYXVΔserp2 intratumorally (n = 5) or post-operatively (n = 5). In dogs treated intratumorally, clinical scores were recorded and tumor biopsies and swabs (from the mouth and virus injection site) were analyzed for viral DNA at multiple time-points. In all dogs, blood, urine, and feces were frequently collected to evaluate organ function, virus distribution, and immune response. No detrimental effects of MYXVΔserp2 treatment were observed in any canine cancer patients. No clinically significant changes in complete blood profiles, serum chemistry analyses, or urinalyses were measured. Viral DNA was isolated from one tumor swab, but viral dissemination was not observed. Anti-MYXV antibodies were occasionally detected. These findings provide needed safety information to advance clinical trials using MYXVΔserp2 to treat patients with cancer.
Collapse
Affiliation(s)
- Amy L MacNeill
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA.
| | - Kristen M Weishaar
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA.
| | - Bernard Séguin
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA.
| | - Barbara E Powers
- Veterinary Diagnostic Laboratories, Colorado State University, Fort Collins, CO 80523, USA.
| |
Collapse
|
5
|
Vargas AJ, Sittadjody S, Thangasamy T, Mendoza EE, Limesand KH, Burd R. Exploiting Tyrosinase Expression and Activity in Melanocytic Tumors. Integr Cancer Ther 2017; 10:328-40. [DOI: 10.1177/1534735410391661] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Melanoma is an aggressive tumor that expresses the pigmentation enzyme tyrosinase. Tyrosinase expression increases during tumorigenesis, which could allow for selective treatment of this tumor type by strategies that use tyrosinase activity. Approaches targeting tyrosinase would involve gene transcription or signal transduction pathways mediated by p53 in a direct or indirect manner. Two pathways are proposed for exploiting tyrosinase expression: ( a) a p53-dependent pathway leading to apoptosis or arrest and ( b) a reactive oxygen species–mediated induction of endoplasmic reticulum stress in p53 mutant tumors. Both strategies could use tyrosinase-mediated activation of quercetin, a dietary polyphenol that induces the expression of p53 and modulates reactive oxygen species. In addition to antitumor signaling properties, activation of quercetin could complement conventional cancer therapy by the induction of phase II detoxification enzymes resulting in p53 stabilization and transduction of its downstream targets. In conclusion, recent advances in tyrosinase enzymology, prodrug chemistry, and modern chemotherapeutics present an intriguing and selective multitherapy targeting system where dietary bioflavonoids could be used to complement conventional cancer treatments.
Collapse
|
6
|
Wang Y, Tai W, Yang J, Zhao G, Sun S, Tseng CTK, Jiang S, Zhou Y, Du L, Gao J. Receptor-binding domain of MERS-CoV with optimal immunogen dosage and immunization interval protects human transgenic mice from MERS-CoV infection. Hum Vaccin Immunother 2017; 13:1615-1624. [PMID: 28277821 DOI: 10.1080/21645515.2017.1296994] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Middle East respiratory syndrome (MERS) continues to raise worldwide concerns due to its pandemic potential. Increased MERS cases and no licensed MERS vaccines highlight the need to develop safe and effective vaccines against MERS. We have previously demonstrated that a receptor-binding domain (RBD) fragment containing residues 377-588 of MERS-coronavirus (MERS-CoV) spike protein is a critical neutralizing domain and an important vaccine target. Nevertheless, its optimal immunogen dosage and immunization interval, key factors for human-used vaccines that induce protective immunity, have never been investigated. In this study, we optimized these criteria using a recombinant MERS-CoV RBD protein fused with Fc (S377-588-Fc) and utilized the optimal immunization schedule to evaluate the protective efficacy of RBD against MERS-CoV infection in human dipeptidyl peptidase 4 transgenic (hDPP4-Tg) mice. Compared with one dose and 2 doses at 1-, 2-, and 3-week intervals, a regimen of 2 doses of this protein separated by an interval of 4 weeks induced the strongest antibody response and neutralizing antibodies against MERS-CoV infection, and maintained at a high level during the detection period. Notably, RBD protein at the optimal dosage and interval protected hDPP4-Tg mice against lethal MERS-CoV challenge, and the protection was positively correlated with serum neutralizing antibodies. Taken together, the optimal immunogen dosage and immunization interval identified in this study will provide useful guidelines for further development of MERS-CoV RBD-based vaccines for human use.
Collapse
Affiliation(s)
- Yufei Wang
- a School of Medical Laboratory Science , Wenzhou Medical University , Wenzhou , Zhejiang , China.,b Lindsley F. Kimball Research Institute , New York Blood Center , New York , NY , USA.,c State Key Laboratory of Pathogen and Biosecurity , Beijing Institute of Microbiology and Epidemiology , Beijing , China
| | - Wanbo Tai
- b Lindsley F. Kimball Research Institute , New York Blood Center , New York , NY , USA.,c State Key Laboratory of Pathogen and Biosecurity , Beijing Institute of Microbiology and Epidemiology , Beijing , China
| | - Jie Yang
- b Lindsley F. Kimball Research Institute , New York Blood Center , New York , NY , USA
| | - Guangyu Zhao
- c State Key Laboratory of Pathogen and Biosecurity , Beijing Institute of Microbiology and Epidemiology , Beijing , China
| | - Shihui Sun
- c State Key Laboratory of Pathogen and Biosecurity , Beijing Institute of Microbiology and Epidemiology , Beijing , China
| | - Chien-Te K Tseng
- d Department of Microbiology and Immunology and Center for Biodefense and Emerging Disease , University of Texas Medical Branch , Galveston , TX , USA
| | - Shibo Jiang
- b Lindsley F. Kimball Research Institute , New York Blood Center , New York , NY , USA.,e Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Basic Medical College and Institute of Medical Microbiology , Fudan University , Shanghai , China
| | - Yusen Zhou
- a School of Medical Laboratory Science , Wenzhou Medical University , Wenzhou , Zhejiang , China.,c State Key Laboratory of Pathogen and Biosecurity , Beijing Institute of Microbiology and Epidemiology , Beijing , China
| | - Lanying Du
- b Lindsley F. Kimball Research Institute , New York Blood Center , New York , NY , USA
| | - Jimin Gao
- a School of Medical Laboratory Science , Wenzhou Medical University , Wenzhou , Zhejiang , China
| |
Collapse
|
7
|
Modified Vaccinia Ankara Virus Vaccination Provides Long-Term Protection against Nasal Rabbitpox Virus Challenge. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2016; 23:648-51. [PMID: 27146001 DOI: 10.1128/cvi.00216-16] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 04/29/2016] [Indexed: 02/08/2023]
Abstract
Modified vaccinia Ankara virus (MVA) is a smallpox vaccine candidate. This study was performed to determine if MVA vaccination provides long-term protection against rabbitpox virus (RPXV) challenge, an animal model of smallpox. Two doses of MVA provided 100% protection against a lethal intranasal RPXV challenge administered 9 months after vaccination.
Collapse
|
8
|
Gene Expression Driven by a Strong Viral Promoter in MVA Increases Vaccination Efficiency by Enhancing Antibody Responses and Unmasking CD8⁺ T Cell Epitopes. Vaccines (Basel) 2014; 2:581-600. [PMID: 26344747 PMCID: PMC4494220 DOI: 10.3390/vaccines2030581] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 06/09/2014] [Accepted: 06/25/2014] [Indexed: 01/11/2023] Open
Abstract
Viral vectors are promising tools for vaccination strategies and immunotherapies. However, CD8+ T cell responses against pathogen-derived epitopes are usually limited to dominant epitopes and antibody responses to recombinant encoded antigens (Ags) are mostly weak. We have previously demonstrated that the timing of viral Ag expression in infected professional Ag-presenting cells strongly shapes the epitope immunodominance hierarchy. T cells recognizing determinants derived from late viral proteins have a clear disadvantage to proliferate during secondary responses. In this work we evaluate the effect of overexpressing the recombinant Ag using the modified vaccinia virus early/late promoter H5 (mPH5). Although the Ag-expression from the natural promoter 7.5 (P7.5) and the mPH5 seemed similar, detailed analysis showed that mPH5 not only induces higher expression levels than P7.5 during early phase of infection, but also Ag turnover is enhanced. The strong overexpression during the early phase leads to broader CD8 T cell responses, while preserving the priming efficiency of stable Ags. Moreover, the increase in Ag-secretion favors the induction of strong antibody responses. Our findings provide the rationale to develop new strategies for fine-tuning the responses elicited by recombinant modified vaccinia virus Ankara by using selected promoters to improve the performance of this viral vector.
Collapse
|
9
|
Hardwick NR, Carroll M, Kaltcheva T, Qian D, Lim D, Leong L, Chu P, Kim J, Chao J, Fakih M, Yen Y, Espenschied J, Ellenhorn JDI, Diamond DJ, Chung V. p53MVA therapy in patients with refractory gastrointestinal malignancies elevates p53-specific CD8+ T-cell responses. Clin Cancer Res 2014; 20:4459-70. [PMID: 24987057 DOI: 10.1158/1078-0432.ccr-13-3361] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE To conduct a phase I trial of a modified vaccinia Ankara (MVA) vaccine delivering wild-type human p53 (p53MVA) in patients with refractory gastrointestinal cancers. EXPERIMENTAL DESIGN Three patients were vaccinated with 1.0×10(8) plaque-forming unit (pfu) p53MVA followed by nine patients at 5.6×10(8) pfu. Toxicity was classified using the NCI Common Toxicity Criteria and clinical responses were assessed by CT scan. Peripheral blood samples were collected pre- and post-immunization for immunophenotyping, monitoring of p53MVA-induced immune response, and examination of PD1 checkpoint inhibition in vitro. RESULTS p53MVA immunization was well tolerated at both doses, with no adverse events above grade 2. CD4+ and CD8+ T cells showing enhanced recognition of a p53 overlapping peptide library were detectable after the first immunization, particularly in the CD8+ T-cell compartment (P=0.03). However, in most patients, this did not expand further with the second and third immunization. The frequency of PD1+ T cells detectable in patients' peripheral blood mononuclear cells (PBMC) was significantly higher than in healthy controls. Furthermore, the frequency of PD1+ CD8+ T cells showed an inverse correlation with the peak CD8+ p53 response (P=0.02) and antibody blockade of PD1 in vitro increased the p53 immune responses detected after the second or third immunizations. Induction of strong T-cell and antibody responses to the MVA backbone were also apparent. CONCLUSION p53MVA was well tolerated and induced robust CD8+ T-cell responses. Combination of p53MVA with immune checkpoint inhibition could help sustain immune responses and lead to enhanced clinical benefit.
Collapse
Affiliation(s)
- Nicola R Hardwick
- Division of Translational Vaccine Research, Beckman Research Institute, City of Hope National Medical Center, Duarte, California
| | - Mary Carroll
- Department of Medical Oncology, City of Hope National Medical Center, Duarte, California
| | - Teodora Kaltcheva
- Division of Translational Vaccine Research, Beckman Research Institute, City of Hope National Medical Center, Duarte, California
| | - Dajun Qian
- Bioinformatics Core Facility, City of Hope National Medical Center, Duarte, California
| | - Dean Lim
- Department of Medical Oncology, City of Hope National Medical Center, Duarte, California
| | - Lucille Leong
- Department of Medical Oncology, City of Hope National Medical Center, Duarte, California
| | - Peiguo Chu
- Department of Pathology, City of Hope National Medical Center, Duarte, California
| | - Joseph Kim
- Department of Surgical Oncology, City of Hope National Medical Center, Duarte, California
| | - Joseph Chao
- Department of Medical Oncology, City of Hope National Medical Center, Duarte, California
| | - Marwan Fakih
- Department of Medical Oncology, City of Hope National Medical Center, Duarte, California
| | - Yun Yen
- Department of Medical Oncology, City of Hope National Medical Center, Duarte, California
| | - Jonathan Espenschied
- Division of Cancer Etiology and Outcomes Research, City of Hope National Medical Center, Duarte, California
| | | | - Don J Diamond
- Division of Translational Vaccine Research, Beckman Research Institute, City of Hope National Medical Center, Duarte, California.
| | - Vincent Chung
- Department of Medical Oncology, City of Hope National Medical Center, Duarte, California
| |
Collapse
|
10
|
Brown EL, Ramiya VK, Wright CA, Jerald MM, Via AD, Kuppala VN, Hazell WS, Lawman PD, Lawman MJ. Treatment of Metastatic Equine Melanoma with a Plasmid DNA Vaccine Encoding Streptococcus Pyogenes EMM55 Protein. J Equine Vet Sci 2014. [DOI: 10.1016/j.jevs.2013.11.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
11
|
Acres B, Bonnefoy JY. Clinical development of MVA-based therapeutic cancer vaccines. Expert Rev Vaccines 2014; 7:889-93. [DOI: 10.1586/14760584.7.7.889] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
12
|
Khan-Farooqi HR, Prins RM, Liau LM. Tumor immunology, immunomics and targeted immunotherapy for central nervous system malignancies. Neurol Res 2013; 27:692-702. [PMID: 16197806 DOI: 10.1179/016164105x49490] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
Although the brain was traditionally considered as 'immunologically privileged', recent findings have implied an involvement of immune mechanisms in neurological disease and illness, including central nervous system (CNS) malignancies. In this review, we initially focus on aspects of the immune system critical for effective antitumor immunity, as an understanding of normal immunological functions and how they relate to tumor immunology will set a foundation for understanding the unique challenges facing the integration of neuro-oncology and neuroimmunology. We summarize current knowledge of immune responses in the 'immunologically quiescent' brain and its role in tumor immunology. We will then discuss the emerging field of 'immunomics' and recent advances in molecular technologies, such as DNA microarray, which are being applied to brain tumor antigen epitope discovery and patient stratification for brain cancer immunotherapy. This, in turn, should have significant importance for ultimately designing and developing efficient and focused strategies for anticancer immunotherapy. Finally, the current state of immune-based treatment paradigms and future directions will be discussed, paying particular attention to targeted antibody strategies, adoptive cellular immunotherapy, and tumor vaccine approaches that have been studied in clinical trials for CNS neoplasms.
Collapse
Affiliation(s)
- Haumith R Khan-Farooqi
- Department of Neurosurgery, David Geffen School of Medicine at UCLA, University of California at Los Angeles, Los Angeles, California 90095-6901, USA
| | | | | |
Collapse
|
13
|
Development of Immunologic Assays to Measure Response in Horses Vaccinated with Xenogeneic Plasmid DNA Encoding Human Tyrosinase. J Equine Vet Sci 2012. [DOI: 10.1016/j.jevs.2012.02.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
14
|
Guzman E, Cubillos-Zapata C, Cottingham MG, Gilbert SC, Prentice H, Charleston B, Hope JC. Modified vaccinia virus Ankara-based vaccine vectors induce apoptosis in dendritic cells draining from the skin via both the extrinsic and intrinsic caspase pathways, preventing efficient antigen presentation. J Virol 2012; 86:5452-66. [PMID: 22419811 PMCID: PMC3347273 DOI: 10.1128/jvi.00264-12] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Accepted: 03/05/2012] [Indexed: 02/03/2023] Open
Abstract
Dendritic cells (DC) are potent antigen-presenting cells and central to the induction of immune responses following infection or vaccination. The collection of DC migrating from peripheral tissues by cannulation of the afferent lymphatic vessels provides DC which can be used directly ex vivo without extensive in vitro manipulations. We have previously used bovine migrating DC to show that recombinant human adenovirus 5 vectors efficiently transduce afferent lymph migrating DEC-205(+) CD11c(+) CD8(-) DC (ALDC). We have also shown that recombinant modified vaccinia virus Ankara (MVA) infects ALDC in vitro, causing downregulation of costimulatory molecules, apoptosis, and cell death. We now show that in the bovine system, modified vaccinia virus Ankara-induced apoptosis in DC draining from the skin occurs soon after virus binding via the caspase 8 pathway and is not associated with viral gene expression. We also show that after virus entry, the caspase 9 pathway cascade is initiated. The magnitude of T cell responses to mycobacterial antigen 85A (Ag85A) expressed by recombinant MVA-infected ALDC is increased by blocking caspase-induced apoptosis. Apoptotic bodies generated by recombinant MVA (rMVA)-Ag85A-infected ALDC and containing Ag85A were phagocytosed by noninfected migrating ALDC expressing SIRPα via actin-dependent phagocytosis, and these ALDC in turn presented antigen. However, the addition of fresh ALDC to MVA-infected cultures did not improve on the magnitude of the T cell responses; in contrast, these noninfected DC showed downregulation of major histocompatibility complex class II (MHC-II), CD40, CD80, and CD86. We also observed that MVA-infected ALDC promoted migration of DEC-205(+) SIRPα(+) CD21(+) DC as well as CD4(+) and CD8(+) T cells independently of caspase activation. These in vitro studies show that induction of apoptosis in DC by MVA vectors is detrimental to the subsequent induction of T cell responses.
Collapse
Affiliation(s)
- E Guzman
- Institute for Animal Health, Compton, Newbury, Berkshire, United Kingdom.
| | | | | | | | | | | | | |
Collapse
|
15
|
Verheust C, Goossens M, Pauwels K, Breyer D. Biosafety aspects of modified vaccinia virus Ankara (MVA)-based vectors used for gene therapy or vaccination. Vaccine 2012; 30:2623-32. [DOI: 10.1016/j.vaccine.2012.02.016] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2011] [Revised: 02/01/2012] [Accepted: 02/05/2012] [Indexed: 11/16/2022]
|
16
|
Tschoep-Lechner K, Drexler I, Hammer D, Neumann D, Pohla H, Sutter G, Noessner E, Issels RD. Modified vaccinia virus Ankara delivers a robust surrogate marker for immune monitoring to sarcoma cells even if cells are being exposed to chemotherapy and heat treatment. Int J Hyperthermia 2012; 28:33-42. [PMID: 22235783 PMCID: PMC9476112 DOI: 10.3109/02656736.2011.626834] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Katharina Tschoep-Lechner
- Department of Internal Medicine III, Klinikum Grosshadern Medical Centre, Ludwig Maximilians University, Marchioninistrasse 25, Munich, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Song GY, Srivastava T, Ishizaki H, Lacey SF, Diamond DJ, Ellenhorn JDI. Recombinant modified vaccinia virus ankara (MVA) expressing wild-type human p53 induces specific antitumor CTL expansion. Cancer Invest 2011; 29:501-10. [PMID: 21843052 PMCID: PMC3260009 DOI: 10.3109/07357907.2011.606248] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The p53 gene product is an attractive target for tumor immunotherapy. The present study aims to understand the potential of MVAp53 vaccine to induce expansion of p53-specific cytotoxic T lymphocyte ex vivo in cancer patients. The result indicated that 14 of 23 cancer patients demonstrated p53-specific IFN-γ production, degranulation, cell proliferation, and lysis of p53 overexpressed human tumor cell lines. These experiments show that MVAp53 stimulation has the potential to induce the expansion of p53-specific cytotoxic T lymphocyte from the memory T cell repertoire. The data suggest that MVAp53 vaccine is an ideal candidate for cancer immunotherapy.
Collapse
Affiliation(s)
- Guang-Yun Song
- Division of Translational Vaccine Research, City of Hope National Medical Center, Duarte, California, USA
| | | | | | | | | | | |
Collapse
|
18
|
Flechsig C, Suezer Y, Kapp M, Tan SM, Löffler J, Sutter G, Einsele H, Grigoleit GU. Uptake of antigens from modified vaccinia Ankara virus-infected leukocytes enhances the immunostimulatory capacity of dendritic cells. Cytotherapy 2011; 13:739-52. [DOI: 10.3109/14653249.2010.549123] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
19
|
Hemmerling J, Wegner-Kops J, von Stebut E, Wolff D, Wagner EM, Hartwig UF, André MC, Theobald M, Schopf RE, Herr W, Meyer RG. Human epidermal Langerhans cells replenish skin xenografts and are depleted by alloreactive T cells in vivo. THE JOURNAL OF IMMUNOLOGY 2011; 187:1142-9. [PMID: 21697461 DOI: 10.4049/jimmunol.1001491] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Epidermal Langerhans cells (LC) are potent APCs surveying the skin. They are crucial regulators of T cell activation in the context of inflammatory skin disease and graft-versus-host disease (GVHD). In contrast to other dendritic cell subtypes, murine LC are able to reconstitute after local depletion without the need of peripheral blood-derived precursors. In this study, we introduce an experimental model of human skin grafted to NOD-SCID IL2Rγ(null) mice. In this model, we demonstrate that xenografting leads to the transient loss of LC from the human skin grafts. Despite the lack of a human hematopoietic system, human LC repopulated the xenografts 6 to 9 wk after transplantation. By staining of LC with the proliferation marker Ki67, we show that one third of the replenishing LC exhibit proliferative activity in vivo. We further used the skin xenograft as an in vivo model for human GVHD. HLA-disparate third-party T cells stimulated with skin donor-derived dendritic cells were injected intravenously into NOD-SCID IL2Rγ(null) mice that had been transplanted with human skin. The application of alloreactive T cells led to erythema and was associated with histological signs of GVHD limited to the transplanted human skin. The inflammation also led to the depletion of LC from the epidermis. In summary, we provide evidence that human LC are able to repopulate the skin independent of blood-derived precursor cells and that this at least partly relates to their proliferative capacity. Our data also propose xeno-transplantation of human skin as a model system for studying the role of skin dendritic cells in the efferent arm of GVHD.
Collapse
Affiliation(s)
- Julia Hemmerling
- Department of Hematology, Oncology, and Pneumology, University Medical Center, Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
DNA vaccination: using the patient's immune system to overcome cancer. Clin Dev Immunol 2010; 2010:169484. [PMID: 21197271 PMCID: PMC3010826 DOI: 10.1155/2010/169484] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2010] [Revised: 10/08/2010] [Accepted: 10/21/2010] [Indexed: 12/15/2022]
Abstract
Cancer is one of the most challenging diseases of today. Optimization of standard treatment protocols consisting of the main columns of chemo- and radiotherapy followed or preceded by surgical intervention is often limited by toxic side effects and induction of concomitant malignancies and/or development of resistant mechanisms. This requires the development of therapeutic strategies which are as effective as standard therapies but permit the patients a life without severe negative side effects. Along this line, the development of immunotherapy in general and the innovative concept of DNA vaccination in particular may provide a venue to achieve this goal. Using the patient's own immune system by activation of humoral and cellular immune responses to target the cancer cells has shown first promising results in clinical trials and may allow reduced toxicity standard therapy regimen in the future. The main challenge of this concept is to transfer the plethora of convincing preclinical and early clinical results to an effective treatment of patients.
Collapse
|
21
|
Nörder M, Becker PD, Drexler I, Link C, Erfle V, Guzmán CA. Modified vaccinia virus Ankara exerts potent immune modulatory activities in a murine model. PLoS One 2010; 5:e11400. [PMID: 20628596 PMCID: PMC2900180 DOI: 10.1371/journal.pone.0011400] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2010] [Accepted: 05/31/2010] [Indexed: 11/18/2022] Open
Abstract
Background Modified vaccinia virus Ankara (MVA), a highly attenuated strain of vaccinia virus, has been used as vaccine delivery vector in preclinical and clinical studies against infectious diseases and malignancies. Here, we investigated whether an MVA which does not encode any antigen (Ag) could be exploited as adjuvant per se. Methodology/Principal Findings We showed that dendritic cells infected in vitro with non-recombinant (nr) MVA expressed maturation and activation markers and were able to efficiently present exogenously pulsed Ag to T cells. In contrast to the dominant T helper (Th) 1 biased responses elicited against Ags produced by recombinant MVA vectors, the use of nrMVA as adjuvant for the co-administered soluble Ags resulted in a long lasting mixed Th1/Th2 responses. Conclusions/Significance These findings open new ways to potentiate and modulate the immune responses to vaccine Ags depending on whether they are co-administered with MVA or encoded by recombinant viruses.
Collapse
Affiliation(s)
- Miriam Nörder
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Pablo D. Becker
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Ingo Drexler
- Institute of Virology, Technische Universität München and Helmholtz Centre Munich, Munich, Germany
| | - Claudia Link
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Volker Erfle
- Institute of Virology, Technische Universität München and Helmholtz Centre Munich, Munich, Germany
- Clinical Cooperation Group Antigen Specific Immunomodulation, Technische Universität München and Helmholtz Centre Munich, Munich, Germany
| | - Carlos A. Guzmán
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
- * E-mail:
| |
Collapse
|
22
|
Manuel ER, Wang Z, Li Z, La Rosa C, Zhou W, Diamond DJ. Intergenic region 3 of modified vaccinia ankara is a functional site for insert gene expression and allows for potent antigen-specific immune responses. Virology 2010; 403:155-62. [PMID: 20471051 DOI: 10.1016/j.virol.2010.04.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2009] [Revised: 12/21/2009] [Accepted: 04/15/2010] [Indexed: 11/16/2022]
Abstract
Integration of exogenous DNA into modified vaccinia Ankara (MVA) is often accomplished using mapped deletion sites in the viral genome. Since MVA has a large capacity (> or =30kb) for foreign gene inserts and a limited number of unique integration sites, development of additional integration sites is needed to take full advantage of the extraordinary capacity for foreign gene insertion. In this report, we evaluate an alternative insertion site known as intergenic region 3 (IGR3). Recombinant MVA carrying the cytomegalovirus pp65 gene in IGR3 (rMVA-pp65-IGR3) demonstrated expression and genetic stability of the insert gene upon passage. Immunization of transgenic HLA-A2 mice with rMVA-pp65-IGR3 induced robust antigen-specific immune responses. Moreover, rMVA-pp65-IGR3-infected human EBV-transformed B cell lines were able to stimulate high levels of pp65-specific memory T cell responses in human PBMCs. These data support the usage of IGR3 for the development of highly immunogenic rMVA vaccines for clinical or veterinary use.
Collapse
Affiliation(s)
- Edwin R Manuel
- Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA
| | | | | | | | | | | |
Collapse
|
23
|
Han SJ, Kaur G, Yang I, Lim M. Biologic Principles of Immunotherapy for Malignant Gliomas. Neurosurg Clin N Am 2010; 21:1-16. [DOI: 10.1016/j.nec.2009.08.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
24
|
Carson C, Antoniou M, Ruiz-Argüello MB, Alcami A, Christodoulou V, Messaritakis I, Blackwell JM, Courtenay O. A prime/boost DNA/Modified vaccinia virus Ankara vaccine expressing recombinant Leishmania DNA encoding TRYP is safe and immunogenic in outbred dogs, the reservoir of zoonotic visceral leishmaniasis. Vaccine 2009; 27:1080-6. [PMID: 19095029 PMCID: PMC2663027 DOI: 10.1016/j.vaccine.2008.11.094] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2008] [Revised: 11/19/2008] [Accepted: 11/27/2008] [Indexed: 01/28/2023]
Abstract
Previous studies demonstrated safety, immunogenicity and efficacy of DNA/modified vaccinia virus Ankara (MVA) prime/boost vaccines expressing tryparedoxin peroxidase (TRYP) and Leishmania homologue of the mammalian receptor for activated C kinase (LACK) against Leishmania major challenge in mice, which was consistent with results from TRYP protein/adjuvant combinations in non-human primates. This study aimed to conduct safety and immunogenicity trials of these DNA/MVA vaccines in dogs, the natural reservoir host of Leishmania infantum, followed-up for 4 months post-vaccination. In a cohort of 22 uninfected outbred dogs, blinded randomised administration of 1000 microg (high dose) or 100 microg (low dose) DNA prime (day 0) and 1x10(8)pfu MVA boost (day 28) was shown to be safe and showed no clinical side effects. High dose DNA/MVA vaccinated TRYP dogs produced statistically higher mean levels of the type-1 pro-inflammatory cytokine IFN-gamma than controls in whole blood assays (WBA) stimulated with the recombinant vaccine antigen TRYP, up to the final sampling at day 126, and in the absence of challenge with Leishmania. TRYP vaccinated dogs also demonstrated significantly higher TRYP-specific total IgG and IgG2 subtype titres than in controls, and positive in vivo intradermal reactions at day 156 in the absence of natural infection, observed in 6/8 TRYP vaccinated dogs. No significant increases in IFN-gamma in LACK-stimulated WBA, or in LACK-specific IgG levels, were detected in LACK vaccinated dogs compared to controls, and only 2/9 LACK vaccinated dogs demonstrated DTH responses at day 156. In all groups, IgG1 subclass responses and antigen-specific stimulation of IL-10 were similar to controls demonstrating an absence of Th2/T(reg) response, as expected in the absence of in vivo restimulation or natural/experimental challenge with Leishmania. These collective results indicate significant antigen-specific type-1 responses and in vivo memory phase cellular immune responses, consistent with superior potential for protective vaccine immunogenicity of DNA/MVA TRYP over LACK.
Collapse
MESH Headings
- Animals
- Antibodies, Protozoan/blood
- Antigens, Protozoan/genetics
- Antigens, Protozoan/immunology
- Dogs
- Immunization, Secondary/methods
- Immunoglobulin G/blood
- Interferon-gamma/metabolism
- Leishmania infantum/immunology
- Leishmaniasis Vaccines/administration & dosage
- Leishmaniasis Vaccines/adverse effects
- Leishmaniasis Vaccines/immunology
- Leishmaniasis, Visceral/immunology
- Leishmaniasis, Visceral/prevention & control
- Leukocytes, Mononuclear/immunology
- Peroxidases/genetics
- Peroxidases/immunology
- Protozoan Proteins/genetics
- Protozoan Proteins/immunology
- Vaccines, DNA/administration & dosage
- Vaccines, DNA/adverse effects
- Vaccines, DNA/immunology
- Vaccinia virus/genetics
Collapse
Affiliation(s)
- Connor Carson
- Populations and Disease Research Group, Department of Biological Sciences, University of Warwick, Gibbet Hill Road, Coventry CV4 7AL, UK.
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Kennedy JS, Greenberg RN. IMVAMUNE: modified vaccinia Ankara strain as an attenuated smallpox vaccine. Expert Rev Vaccines 2009; 8:13-24. [PMID: 19093767 PMCID: PMC9709931 DOI: 10.1586/14760584.8.1.13] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Smallpox vaccines based on replicating vaccinia virus are known to elicit rare yet serious adverse events, particularly in human populations with immune deficiency, atopic dermatitis and at the extremes of age. A vaccine that induces protective immune responses equivalent to first-generation smallpox vaccines while reducing the risk for severe adverse events is critical for a national stockpile of smallpox vaccines. Modified vaccinia Ankara (MVA) has been proposed as an immediate solution for vaccination of high-risk individuals. Bavarian Nordic's vaccine MVA-BN (IMVAMUNE) is a MVA strain that is replication incompetent in mammalian cell lines. IMVAMUNE has been administered to more than 1900 human subjects to date, including high-risk populations (e.g., people diagnosed with atopic dermatitis or infected with HIV) in which standard replicating vaccines are contraindicated. We review the Phase I clinical trial safety profile and immune responses and compare them with other smallpox vaccines, including ACAM2000 and Dryvax.
Collapse
Affiliation(s)
- Jeffrey S Kennedy
- Division of Infectious Diseases, Wadsworth Center, NYS Department of Health, Biggs Laboratory, C606, PO Box 509, Albany, NY 12201-0509, USA.
| | - Richard N Greenberg
- VA Staff Physician, Lexington VA Medical Center and Professor of Medicine, The Belinda Mason Carden and Paul Mason Professor of HIV/AIDS Research and Education, University of Kentucky School of Medicine, Department of Medicine, Room MN-672, 800 Rose Street, Lexington, KY 40536-0084, USA.
| |
Collapse
|
26
|
Nino-Fong R, Johnston JB. Poxvirus-based vaccine platforms: getting at those hard-to-reach places. Future Virol 2008. [DOI: 10.2217/17460794.3.2.99] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Affiliation(s)
- Rodolfo Nino-Fong
- Institute for Nutrisciences & Health, National Research Council Canada, 550 University Avenue, Charlottetown, PE, C1A 4P3, Canada
| | - James B Johnston
- Institute for Nutrisciences & Health, National Research Council Canada, 550 University Avenue, Charlottetown, PE, C1A 4P3, Canada
| |
Collapse
|
27
|
Burgers WA, Shephard E, Monroe JE, Greenhalgh T, Binder A, Hurter E, Van Harmelen JH, Williamson C, Williamson AL. Construction, characterization, and immunogenicity of a multigene modified vaccinia Ankara (MVA) vaccine based on HIV type 1 subtype C. AIDS Res Hum Retroviruses 2008; 24:195-206. [PMID: 18240957 DOI: 10.1089/aid.2007.0205] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Candidate vaccines composed of a DNA construct to prime the immune system, followed by modified vaccinia Ankara (MVA) containing matching genes as a booster vaccination, have produced encouraging immune responses in human volunteers. This study presents the detailed construction and characterization of a recombinant MVA that will be tested in combination with a DNA vaccine in Phase I clinical trials in South Africa and the United States. To match recently transmitted viruses in the southern African region and to maximize epitope coverage, the vaccines were constructed to contain five HIV-1 subtype C genes, namely gag, reverse transcriptase, tat, and nef (grttn), expressed as a polyprotein, and a truncated env (gp150). An initial recombinant MVA construct containing wild-type env was found to be genetically unstable, and thus a human codon-optimized gene was used. Grttn and gp150 were inserted into two different sites in MVA yielding a double recombinant, SAAVI MVA-C. The recombinant MVA was shown to be genetically stable and high level expression of the transgenes was observed. Env retained infectivity in a functional infectivity assay despite a point mutation that arose during virus generation. Mice inoculated with SAAVI MVA-C at various doses developed high levels of Gag, RT, and Env-specific CD8(+) and CD4(+) T cells, and some of these responses could be boosted by a second inoculation. An accompanying paper describes the immunogenicity of SAAVI MVA-C when given in combination with SAAVI DNA-C.
Collapse
Affiliation(s)
- Wendy A. Burgers
- Institute of Infectious Disease and Molecular Medicine and Division of Medical Virology, Department of Clinical Laboratory Sciences, University of Cape Town, Cape Town, South Africa
| | - Enid Shephard
- Institute of Infectious Disease and Molecular Medicine and Division of Medical Virology, Department of Clinical Laboratory Sciences, University of Cape Town, Cape Town, South Africa
- MRC/UCT Liver Research Centre, University of Cape Town, Cape Town, South Africa
| | | | | | - Anke Binder
- Institute of Infectious Disease and Molecular Medicine and Division of Medical Virology, Department of Clinical Laboratory Sciences, University of Cape Town, Cape Town, South Africa
| | - Etienne Hurter
- Institute of Infectious Disease and Molecular Medicine and Division of Medical Virology, Department of Clinical Laboratory Sciences, University of Cape Town, Cape Town, South Africa
| | - Joanne H. Van Harmelen
- Institute of Infectious Disease and Molecular Medicine and Division of Medical Virology, Department of Clinical Laboratory Sciences, University of Cape Town, Cape Town, South Africa
| | - Carolyn Williamson
- Institute of Infectious Disease and Molecular Medicine and Division of Medical Virology, Department of Clinical Laboratory Sciences, University of Cape Town, Cape Town, South Africa
| | - Anna-Lise Williamson
- Institute of Infectious Disease and Molecular Medicine and Division of Medical Virology, Department of Clinical Laboratory Sciences, University of Cape Town, Cape Town, South Africa
- National Health Laboratory Services, Groote Schuur Hospital, Cape Town, South Africa
| |
Collapse
|
28
|
Huang X, Liu L, Ren L, Qiu C, Wan Y, Xu J. Mucosal priming with replicative Tiantan vaccinia and systemic boosting with DNA vaccine raised strong mucosal and systemic HIV-specific immune responses. Vaccine 2007; 25:8874-84. [DOI: 10.1016/j.vaccine.2007.08.066] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2007] [Revised: 08/21/2007] [Accepted: 08/27/2007] [Indexed: 10/22/2022]
|
29
|
Gasteiger G, Kastenmuller W, Ljapoci R, Sutter G, Drexler I. Cross-priming of cytotoxic T cells dictates antigen requisites for modified vaccinia virus Ankara vector vaccines. J Virol 2007; 81:11925-36. [PMID: 17699574 PMCID: PMC2168793 DOI: 10.1128/jvi.00903-07] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Recombinant vaccines based on modified vaccinia virus Ankara (MVA) have an excellent record concerning safety and immunogenicity and are currently being evaluated in numerous clinical studies for immunotherapy of infectious diseases and cancer. However, knowledge about the biological properties of target antigens to efficiently induce MVA vaccine-mediated immunity in vivo is sparse. Here, we examined distinct antigen presentation pathways and different antigen formulations contained in MVA vaccines for their capability to induce cytotoxic CD8(+) T-cell (CTL) responses. Strikingly, we found that CTL responses against MVA-produced antigens were dominated by cross-priming in vivo, despite the ability of the virus to efficiently infect professional antigen-presenting cells such as dendritic cells. Moreover, stable mature protein was preferred to preprocessed antigen as the substrate for cross-priming. Our data are essential for improved MVA vaccine design, as they demonstrate the need for optimal adjustment of the target antigen properties to the intrinsic requirements of the delivering vector system.
Collapse
Affiliation(s)
- Georg Gasteiger
- GSF-Institute for Molecular Virology, Schneckenburgerstrasse 8, D-81675 Munich, Germany
| | | | | | | | | |
Collapse
|
30
|
Pedersen AE, Ronchese F. CTLA-4 blockade during dendritic cell based booster vaccination influences dendritic cell survival and CTL expansion. JOURNAL OF IMMUNE BASED THERAPIES AND VACCINES 2007; 5:9. [PMID: 17662155 PMCID: PMC1950502 DOI: 10.1186/1476-8518-5-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2007] [Accepted: 07/29/2007] [Indexed: 11/10/2022]
Abstract
Dendritic cells (DCs) are potent antigen-presenting cells and critical for the priming of CD8+ T cells. Therefore the use of these cells as adjuvant cells has been tested in a large number of experimental and clinical vaccination studies, in particular cancer vaccine studies. A number of protocols are emerging that combine vaccination with CTL expanding strategies, such as e.g. blockade of CTLA-4 signalling. On the other hand, the lifespan and in vivo survival of therapeutic DCs have only been addressed in a few studies, although this is of importance for the kinetics of CTL induction during vaccination. We have previously reported that DCs loaded with specific antigens are eliminated by antigen specific CTLs in vivo and that this elimination affects the potential for in vivo CTL generation. We now show that CTLA-4 blockade increases the number of DC vaccine induced LCMV gp33 specific CTLs and the lysis of relevant in vivo targets. However, the CTLA-4 blockage dependent expansion of CTLs also affect DC survival during booster DC injections and our data suggest that during a booster DC vaccine, the largest increase in CTL levels is already obtained during the first vaccination.
Collapse
Affiliation(s)
- Anders E Pedersen
- Department of International Health, Immunology and Microbiology, The Panum Institute, University of Copenhagen, Denmark
| | - Franca Ronchese
- Malaghan Institute of Medical Research, Wellington, New Zealand
| |
Collapse
|
31
|
Meyer RG, Korn S, Micke P, Becker K, Huber C, Wölfel T, Buhl R. An open-label, prospective phase I/II study evaluating the immunogenicity and safety of a ras peptide vaccine plus GM-CSF in patients with non-small cell lung cancer. Lung Cancer 2007; 58:88-94. [PMID: 17599645 DOI: 10.1016/j.lungcan.2007.05.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2007] [Revised: 05/03/2007] [Accepted: 05/13/2007] [Indexed: 01/08/2023]
Abstract
Mutations of the ras gene have been reported in 20-40% of NSCLC patients. If present, they are critical for the malignant phenotype of these tumors. Therefore, targeting them by specific vaccination is a promising therapeutic approach. In a clinical trial we screened for ras mutations in patients with NSCLC. Patients with ras-positive tumors were immunized six times intradermally with a mixture of seven peptides representing the most common ras mutations. Objectives of the study were the feasibility, efficacy and safety of the vaccination. In addition, the induction of a specific immune reaction was investigated by DTH tests, and the induction of peptide-specific T cells was tested in ex vivo IFN-gamma-ELISPOT assays. Five of 18 patients had ras mutations at codon 12. Four of these patients, all with adenocarcinomas (stage I: n=3, stage IV: n=1) entered the study. The patient with stage IV disease withdrew prematurely after the third application because of disease progression associated with pulmonary embolism. Ras-specific T cells were not detected ex vivo. However, one patient developed a positive DTH reaction after the fifth vaccination that increased after the sixth vaccination. Our results are in line with earlier trials reporting ras mutations in 20-40% of NSCLC patients. Vaccination with mutated ras peptides is feasible and well tolerated. One patient revealed a positive DTH test. An ex vivo detectable T cell response was not induced in any of the patients.
Collapse
Affiliation(s)
- Ralf G Meyer
- III. Medical Department, Mainz University Hospital, Langenbeckstrasse 1, 55101 Mainz, Germany
| | | | | | | | | | | | | |
Collapse
|
32
|
Anderson RJ, Schneider J. Plasmid DNA and viral vector-based vaccines for the treatment of cancer. Vaccine 2007; 25 Suppl 2:B24-34. [PMID: 17698262 DOI: 10.1016/j.vaccine.2007.05.030] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2007] [Revised: 05/06/2007] [Accepted: 05/15/2007] [Indexed: 12/14/2022]
Abstract
Plasmid DNA and viral vector-based cancer vaccines have many inherent features that make them promising cancer vaccine candidates. This review focuses on the use of plasmid DNA and viral vector vaccines to deliver tumour-specific antigens to induce a tumour-specific immune response. Examples of different antigen delivery systems that have been tested in recent clinical trials are summarised and advantages and disadvantages of a number of delivery systems and approaches are discussed. Finally, an outlook on how plasmid DNA and viral vectors might be developed further as cancer vaccines is provided.
Collapse
Affiliation(s)
- Richard J Anderson
- Oxxon Therapeutics Ltd., 2nd Floor Florey House, 3 Robert Robinson Avenue, Oxford Science Park, Oxford OX4 4GP, UK
| | | |
Collapse
|
33
|
Meyer RG, Graf C, Britten CM, Huber C, Wölfel T. Rapid identification of an HLA-B*1501-restricted vaccinia peptide antigen. Vaccine 2007; 25:4715-22. [PMID: 17499403 DOI: 10.1016/j.vaccine.2007.04.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2006] [Revised: 03/21/2007] [Accepted: 04/02/2007] [Indexed: 11/25/2022]
Abstract
We identified a modified vaccinia virus Ankara (MVA)-peptide recognized by T cells of a vaccinated patient, applying a modified expression-based procedure. CD8(+) T cells reactive with MVA were expanded from peripheral blood of a patient after MVA-vaccination. The restricting HLA-elements and the antigen were determined applying stably HLA-class I-transfected K562 cells that were either infected wit MVA or transfected with a panel of MVA open reading frames (ORF). The T cells recognized the ORF 28R (K7R) gene product in association with HLA-B*1501 and the peptide-coding region was localized applying truncated in vitro transcribed mRNA. The 9-mer peptide encoded (ORF 28(25-33), SIIDLIDEY) was identified that was also recognized by ex vivo CD8(+) T cells from the patient and further healthy individuals.
Collapse
Affiliation(s)
- Ralf G Meyer
- III Medizinische Klinik, Johannes Gutenberg-Universitaet, Mainz, Germany.
| | | | | | | | | |
Collapse
|
34
|
Wang Z, La Rosa C, Li Z, Ly H, Krishnan A, Martinez J, Britt WJ, Diamond D. Vaccine properties of a novel marker gene-free recombinant modified vaccinia Ankara expressing immunodominant CMV antigens pp65 and IE1. Vaccine 2007; 25:1132-41. [PMID: 17049414 PMCID: PMC1852509 DOI: 10.1016/j.vaccine.2006.09.067] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2006] [Revised: 08/17/2006] [Accepted: 09/13/2006] [Indexed: 11/21/2022]
Abstract
CMV tegument protein pp65 and CMV immediate early gene product IE1 are both considered immunodominant targets of cell-mediated immunity (CMI) and potentially capable of controlling CMV infection. To better assess their role in host defense, we have constructed a novel MVA transfer vector named pZWIIA and generated a recombinant MVA (rMVA) expressing both full-length pp65 and exon4 of IE1 (pp65-IE1-MVA) at high levels, followed by the genetic removal of the bacterial marker gene used to distinguish recombinant forms. Immunogenicity evaluation indicates that pp65-IE1-MVA not only can induce robust primary CMI to both antigens in HLA A2.1 Tg mice, but also can stimulate vigorous expansion of memory T lymphocyte responses to pp65 and IE1 in PBMC of CMV-positive donors. These properties make the MVA-based vaccine ideal for the dual role of priming and boosting CMV-specific T cell immunity as a means to control CMV disease in recipients of hematopoietic cell or solid organ transplantation (HCT or SOT). pZWIIA alone or in combination with other MVA transfer vectors can be used to generate MVA based multiple-antigen vaccine which have application in vaccine development for a wide spectrum of infectious diseases and cancer.
Collapse
Affiliation(s)
- Zhongde Wang
- Laboratory of Vaccine Research, Division of Virology, Beckman Research Institute of City of Hope, Duarte, CA 91010
| | - Corinna La Rosa
- Laboratory of Vaccine Research, Division of Virology, Beckman Research Institute of City of Hope, Duarte, CA 91010
| | - Zhongqi Li
- Laboratory of Vaccine Research, Division of Virology, Beckman Research Institute of City of Hope, Duarte, CA 91010
| | - Heang Ly
- Laboratory of Vaccine Research, Division of Virology, Beckman Research Institute of City of Hope, Duarte, CA 91010
| | - Aparna Krishnan
- Laboratory of Vaccine Research, Division of Virology, Beckman Research Institute of City of Hope, Duarte, CA 91010
| | - Joy Martinez
- Laboratory of Vaccine Research, Division of Virology, Beckman Research Institute of City of Hope, Duarte, CA 91010
| | - William J. Britt
- Department of Pediatrics, University of Alabama, Birmingham, Alabama 352333
| | - Don Diamond
- Laboratory of Vaccine Research, Division of Virology, Beckman Research Institute of City of Hope, Duarte, CA 91010
| |
Collapse
|
35
|
Greiner S, Humrich JY, Thuman P, Sauter B, Schuler G, Jenne L. The highly attenuated vaccinia virus strain modified virus Ankara induces apoptosis in melanoma cells and allows bystander dendritic cells to generate a potent anti-tumoral immunity. Clin Exp Immunol 2007; 146:344-53. [PMID: 17034588 PMCID: PMC1942054 DOI: 10.1111/j.1365-2249.2006.03177.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Vaccinia virus (VV) has been tested as oncolytic virus against malignant melanoma in clinical trials for more than 40 years. Until now, mainly strains comparable to viral strains used for smallpox vaccination have been probed for anti-tumoral therapy. We have shown recently that the wild-type strain Western Reserve (WR) can interfere with crucial functions of monocyte-derived dendritic cells (DCs). Our aim was to examine whether viral immune evasion mechanisms might be responsible for the ineffectiveness of WR-based vaccination strategies and whether the highly attenuated strain modified virus Ankara (MVA) differs from WR with respect to its possible immunostimulatory capacity after intratumoral injection. Using in vitro experiments, we compared the effect of both strains on melanoma cells and on local bystander DCs. We found that both VV-strains infected melanoma cells efficiently and caused disintegration of the actin cytoskeleton, as shown by fluorescence microscopy. In addition, both VV-strains caused apoptotic cell death in melanoma cells after infection. In contrast to MVA, WR underwent a complete viral replication cycle in melanoma cells. Bystander DCs were consecutively infected by newly generated WR virions and lost their capacity to induce allogeneic T cell proliferation. DCs in contact with MVA-infected melanoma cells retained their capacity to induce T cell proliferation. Immature DCs were capable of phagocytosing MVA-infected melanoma cells. Priming of autologous CD8(+) T cells by DCs that had phagocytosed MVA-infected, MelanA positive melanoma cells resulted in the induction of T cell clones specifically reactive against the model antigen MelanA as shown by enzyme-linked immunospot (ELISPOT) analysis. We conclude that the clinical trials with oncolytic wild-type VV failed probably because of suppression of bystander DCs and consecutive suppression of T cell-mediated anti-melanoma immunity. The attenuated VV-strain MVA facilitates the generation of tumour associated antigen (TAA)-specific T cell response as it is oncolytic for melanoma cells, but non-toxic for DC, and should be a promising candidate for intralesional metastatic melanoma therapy.
Collapse
Affiliation(s)
- S Greiner
- Department of Dermatology, University Hospital Erlangen, Erlangen, Germany
| | | | | | | | | | | |
Collapse
|
36
|
Zhu J, Martinez J, Huang X, Yang Y. Innate immunity against vaccinia virus is mediated by TLR2 and requires TLR-independent production of IFN-beta. Blood 2006; 109:619-25. [PMID: 16973959 PMCID: PMC1785085 DOI: 10.1182/blood-2006-06-027136] [Citation(s) in RCA: 159] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Vaccinia virus (VV) has been used extensively as a vaccine vehicle in the clinical application for infectious diseases and cancer. Previous studies have suggested that the unique potency of VV-based vaccine lies in its effective activation of the innate immune system. However, how VV activates innate immune pathways remains largely unknown. In this study, we showed that VV elicited innate immune response through both Toll-like receptor (TLR)-dependent and -independent pathways. The TLR pathway was mediated by TLR2 and MyD88, leading to the production of proinflammatory cytokines, whereas activation of the TLR-independent pathway resulted in the secretion of IFN-beta. More importantly, both TLR-dependent and -independent pathways were required for activating innate and adaptive immunity to VV in vivo. These findings represent the first evidence that innate immune recognition of VV is mediated by TLR2, demonstrate that one pathogen can target both TLR and non-TLR innate immune pathways to work together in achieving efficient activation of host defense, and suggest potential new strategies for the design of effective vaccines.
Collapse
Affiliation(s)
- Jiangao Zhu
- Department of Medicine, Division of Medical Oncology, and
| | - Jennifer Martinez
- Department of Immunology, Duke University Medical Center, Durham, NC
| | - Xiaopei Huang
- Department of Medicine, Division of Medical Oncology, and
| | - Yiping Yang
- Department of Medicine, Division of Medical Oncology, and
- Department of Immunology, Duke University Medical Center, Durham, NC
- Correspondence: Yiping Yang,
Departments of Medicine and Immunology, Duke University Medical Center, Box 3502, Durham, NC 27710; e-mail:
| |
Collapse
|
37
|
Hengge UR. Gene therapy progress and prospects: the skin – easily accessible, but still far away. Gene Ther 2006; 13:1555-63. [PMID: 16957767 DOI: 10.1038/sj.gt.3302855] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Significant progress has been made in corrective gene therapy of inherited skin diseases. This includes advances in vector technology, targeted gene expression, gene replacement, and the availability of appropriate animal models for a variety of candidate diseases. In addition, an increased understanding of the uptake and trafficking mechanisms inside keratinocytes has evolved. Topical application facilitates DNA vaccination through the skin, albeit clinical benefits have not yet materialized. However, the translation into clinical trials has only been partially mastered. The latter and the control of immune responses represent challenges for the research community.
Collapse
Affiliation(s)
- U R Hengge
- Department of Dermatology, Heinrich-Heine-University, Duesseldorf, Germany.
| |
Collapse
|
38
|
Chavan R, Marfatia KA, An IC, Garber DA, Feinberg MB. Expression of CCL20 and granulocyte-macrophage colony-stimulating factor, but not Flt3-L, from modified vaccinia virus ankara enhances antiviral cellular and humoral immune responses. J Virol 2006; 80:7676-87. [PMID: 16840346 PMCID: PMC1563727 DOI: 10.1128/jvi.02748-05] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2005] [Accepted: 05/09/2006] [Indexed: 11/20/2022] Open
Abstract
While modified vaccinia virus Ankara (MVA) is currently in clinical development as a safe vaccine against smallpox and heterologous infectious diseases, its immunogenicity is likely limited due to the inability of the virus to replicate productively in mammalian hosts. In light of recent data demonstrating that vaccinia viruses, including MVA, preferentially infect antigen-presenting cells (APCs) that play crucial roles in generating antiviral immunity, we hypothesized that expression of specific cytokines and chemokines that mediate APC recruitment and activation from recombinant MVA (rMVA) vectors would enhance the immunogenicity of these vectors. To test this hypothesis, we generated rMVAs that express murine granulocyte-macrophage colony-stimulating factor (mGM-CSF), human CCL20/human macrophage inflammatory protein 3alpha (hCCL20/hMIP-3alpha), or human fms-like tyrosine kinase 3 ligand (hFlt3-L), factors predicted to increase levels of dendritic cells (DCs), to recruit DCs to sites of immunization, or to promote maturation of DCs in vivo, respectively. These rMVAs also coexpress the well-characterized, immunodominant lymphocytic choriomeningitis virus nucleoprotein (NP) antigen that enabled sensitive and quantitative assessment of antigen-specific CD8(+) T-cell responses following immunization of BALB/c mice. Our results demonstrate that immunization of mice with rMVAs expressing mGM-CSF or hCCL20, but not hFlt3-L, results in two- to fourfold increases of cellular immune responses directed against vector-encoded antigens and 6- to 17-fold enhancements of MVA-specific antibody titers, compared to those responses elicited by nonadjuvanted rMVA. Of note, cytokine augmentation of cellular immune responses occurs when rMVAs are given as primary immunizations but not when they are used as booster immunizations, suggesting that these APC-modulating proteins, when used as poxvirus-encoded adjuvants, are more effective at stimulating naïve T-cell responses than in promoting recall of preexisting memory T-cell responses. Our results demonstrate that a strategy to express specific genetic adjuvants from rMVA vectors can be successfully applied to enhance the immunogenicity of MVA-based vaccines.
Collapse
Affiliation(s)
- R Chavan
- Emory University Vaccine Center, 954 Gatewood Road NE, Atlanta, GA 30329, USA
| | | | | | | | | |
Collapse
|
39
|
Kastenmuller W, Drexler I, Ludwig H, Erfle V, Peschel C, Bernhard H, Sutter G. Infection of human dendritic cells with recombinant vaccinia virus MVA reveals general persistence of viral early transcription but distinct maturation-dependent cytopathogenicity. Virology 2006; 350:276-88. [PMID: 16595141 DOI: 10.1016/j.virol.2006.02.039] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2005] [Revised: 12/27/2005] [Accepted: 02/24/2006] [Indexed: 11/21/2022]
Abstract
Vector-infected dendritic cells (DC) are evaluated for antigen delivery in experimental therapy of cancer and infectious diseases. Here, we investigated infections of immature or mature, monocyte-derived human DC with recombinant vaccinia virus MVA producing human Her-2/neu, a candidate tumor-associated antigen. Assessment of the molecular virus life cycle in infected DC revealed a general arrest at the level of viral early gene expression. When monitoring the phenotype of MVA-infected DC, including expression of cell surface markers, we found immature cells readily undergoing apoptosis. Nevertheless, we detected significant populations of viable DC being characterized by high level Her-2/neu expression and unimpaired display of costimulatory molecules. While infected viable immature DC failed to undergo maturation despite cytokine treatment, both DC populations efficiently presented MVA-produced target antigen. These findings allow to better define the requirements for MVA-mediated antigen delivery to DC and help to derive optimized vectors for this advanced therapy option.
Collapse
Affiliation(s)
- Wolfgang Kastenmuller
- GSF-Institut für Molekulare Virologie, Klinikum rechts der Isar, Technische Universität 81675 München, Germany
| | | | | | | | | | | | | |
Collapse
|
40
|
Pérez-Jiménez E, Kochan G, Gherardi MM, Esteban M. MVA-LACK as a safe and efficient vector for vaccination against leishmaniasis. Microbes Infect 2006; 8:810-22. [PMID: 16504562 DOI: 10.1016/j.micinf.2005.10.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2005] [Revised: 09/15/2005] [Accepted: 10/03/2005] [Indexed: 10/25/2022]
Abstract
An optimal vaccine against leishmaniasis should elicit parasite specific CD4+ and cytotoxic CD8+ T cells. In this investigation, we described a prime/boost immunization approach based on DNA and on poxvirus vectors (Western Reserve, WR, and the highly attenuated modified vaccinia virus Ankara, MVA), both expressing the LACK antigen of Leishmania infantum, that triggers different levels of specific CD8+ T cell responses and protection (reduction in lesion size and parasitemia) against L. major infection in mice. A prime/boost vaccination with DNA-LACK/MVA-LACK elicits higher CD8+ T cell responses than a similar protocol with the replication competent VV-LACK. Both CD4+ and CD8+ T cells were induced by DNA-LACK/MVA-LACK immunization. The levels of IFN-gamma and TNF-alpha secreting CD8+ T cells were higher in splenocytes from DNA-LACK/MVA-LACK than in DNA-LACK/VV-LACK immunized animals. Moreover, protection against L. major was significantly higher in DNA-LACK/MVA-LACK than in DNA-LACK/VV-LACK immunized animals when boosted with the same virus dose, and correlated with high levels of IFN-gamma and TNF-alpha secreting CD8+ T cells. In DNA-LACK/MVA-LACK vaccinated animals, the extent of lesion size reduction ranged from 65 to 92% and this protection was maintained for at least 17 weeks after challenge with the parasite. These findings demonstrate that in heterologous prime/boost immunization approaches, the protocol DNA-LACK/MVA-LACK is superior to DNA-LACK/VV-LACK in triggering specific CD8+ T cell immune responses and in conferring protection against cutaneous leishmaniasis. Thus, MVA-LACK is a safe and efficient vector for vaccination against leishmaniasis.
Collapse
MESH Headings
- Animals
- Antigens, Protozoan/genetics
- Antigens, Protozoan/immunology
- DNA, Protozoan
- Drug Administration Schedule
- Female
- Immunization, Secondary
- Interferon-gamma/metabolism
- Leishmania major
- Leishmaniasis, Cutaneous/immunology
- Leishmaniasis, Cutaneous/pathology
- Leishmaniasis, Cutaneous/prevention & control
- Mice
- Mice, Inbred BALB C
- Protozoan Proteins/genetics
- Protozoan Proteins/immunology
- Protozoan Vaccines/adverse effects
- Protozoan Vaccines/genetics
- Protozoan Vaccines/immunology
- T-Lymphocytes/metabolism
- Tumor Necrosis Factor-alpha/metabolism
- Vaccines, Synthetic/adverse effects
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
- Vaccinia virus
Collapse
Affiliation(s)
- Eva Pérez-Jiménez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, CSIC, Ciudad Universitaria de Cantoblanco, 28049, Madrid, Spain
| | | | | | | |
Collapse
|