1
|
Edwardson TGW, Levasseur MD, Tetter S, Steinauer A, Hori M, Hilvert D. Protein Cages: From Fundamentals to Advanced Applications. Chem Rev 2022; 122:9145-9197. [PMID: 35394752 DOI: 10.1021/acs.chemrev.1c00877] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Proteins that self-assemble into polyhedral shell-like structures are useful molecular containers both in nature and in the laboratory. Here we review efforts to repurpose diverse protein cages, including viral capsids, ferritins, bacterial microcompartments, and designed capsules, as vaccines, drug delivery vehicles, targeted imaging agents, nanoreactors, templates for controlled materials synthesis, building blocks for higher-order architectures, and more. A deep understanding of the principles underlying the construction, function, and evolution of natural systems has been key to tailoring selective cargo encapsulation and interactions with both biological systems and synthetic materials through protein engineering and directed evolution. The ability to adapt and design increasingly sophisticated capsid structures and functions stands to benefit the fields of catalysis, materials science, and medicine.
Collapse
Affiliation(s)
| | | | - Stephan Tetter
- Laboratory of Organic Chemistry, ETH Zurich, 8093 Zurich, Switzerland
| | - Angela Steinauer
- Laboratory of Organic Chemistry, ETH Zurich, 8093 Zurich, Switzerland
| | - Mao Hori
- Laboratory of Organic Chemistry, ETH Zurich, 8093 Zurich, Switzerland
| | - Donald Hilvert
- Laboratory of Organic Chemistry, ETH Zurich, 8093 Zurich, Switzerland
| |
Collapse
|
2
|
Wang X, Diamond DJ, Forman SJ, Nakamura R. Development of CMV-CD19 bi-specific CAR T cells with post-infusion in vivo boost using an anti-CMV vaccine. Int J Hematol 2021; 114:544-553. [PMID: 34561840 PMCID: PMC8475363 DOI: 10.1007/s12185-021-03215-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 08/21/2021] [Accepted: 09/01/2021] [Indexed: 11/25/2022]
Abstract
Adoptive transfer of in vitro expanded, chimeric antigen receptor (CAR)-redirected CD19-specific T cells can induce dramatic disease regression in patients with leukemia and lymphomas. However, the full potential of this emerging modality is hampered in some cancer settings by a significant rate of therapeutic failure arising from the attenuated engraftment and persistence of CAR-redirected T cells, and tumor relapse following adoptive transfer. Here, we discuss an advanced strategy that facilitates post-infusion in vivo boosting of CAR T cells via CMV vaccination, to mediate durable remission of B cell malignancies by engrafting a CAR molecule onto a CMV-specific T cell. We also discuss a feasible and unique platform for the generation of the CMV-CD19CAR T cells for clinical application. This new approach would overcome multiple challenges in current CAR T cell technology including: short T cell persistence, limited duration of response, and inability to re-stimulate T cells after relapse or persistent disease.
Collapse
Affiliation(s)
- Xiuli Wang
- Department of Hematology/HCT, City of Hope Comprehensive Cancer Center and the Beckman Research Institute of City of Hope, 1500 E. Duarte Road, Duarte, CA, 91010, USA
| | - Don J Diamond
- Department of Hematology/HCT, City of Hope Comprehensive Cancer Center and the Beckman Research Institute of City of Hope, 1500 E. Duarte Road, Duarte, CA, 91010, USA
| | - Stephen J Forman
- Department of Hematology/HCT, City of Hope Comprehensive Cancer Center and the Beckman Research Institute of City of Hope, 1500 E. Duarte Road, Duarte, CA, 91010, USA
| | - Ryotaro Nakamura
- Department of Hematology/HCT, City of Hope Comprehensive Cancer Center and the Beckman Research Institute of City of Hope, 1500 E. Duarte Road, Duarte, CA, 91010, USA.
| |
Collapse
|
3
|
Demchuk AM, Patel TR. The biomedical and bioengineering potential of protein nanocompartments. Biotechnol Adv 2020; 41:107547. [PMID: 32294494 DOI: 10.1016/j.biotechadv.2020.107547] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Revised: 03/21/2020] [Accepted: 04/03/2020] [Indexed: 12/18/2022]
Abstract
Protein nanocompartments (PNCs) are self-assembling biological nanocages that can be harnessed as platforms for a wide range of nanobiotechnology applications. The most widely studied examples of PNCs include virus-like particles, bacterial microcompartments, encapsulin nanocompartments, enzyme-derived nanocages (such as lumazine synthase and the E2 component of the pyruvate dehydrogenase complex), ferritins and ferritin homologues, small heat shock proteins, and vault ribonucleoproteins. Structural PNC shell proteins are stable, biocompatible, and tolerant of both interior and exterior chemical or genetic functionalization for use as vaccines, therapeutic delivery vehicles, medical imaging aids, bioreactors, biological control agents, emulsion stabilizers, or scaffolds for biomimetic materials synthesis. This review provides an overview of the recent biomedical and bioengineering advances achieved with PNCs with a particular focus on recombinant PNC derivatives.
Collapse
Affiliation(s)
- Aubrey M Demchuk
- Department of Neuroscience, University of Lethbridge, 4401 University Drive West, Lethbridge, AB, Canada.
| | - Trushar R Patel
- Alberta RNA Research and Training Institute, Department of Chemistry and Biochemistry, University of Lethbridge, 4401 University Drive West, Lethbridge, AB, Canada; Department of Microbiology, Immunology and Infectious Diseases, Cumming, School of Medicine, University of Calgary, 2500 University Dr. N.W., Calgary, AB T2N 1N4, Canada; Li Ka Shing Institute of Virology and Discovery Lab, Faculty of Medicine & Dentistry, University of Alberta, 6-010 Katz Center for Health Research, Edmonton, AB T6G 2E1, Canada.
| |
Collapse
|
4
|
Mehrizi AA, Jafari Zadeh A, Zakeri S, Djadid ND. Population genetic structure analysis of thrombospondin-related adhesive protein (TRAP) as a vaccine candidate antigen in worldwide Plasmodium falciparum isolates. INFECTION GENETICS AND EVOLUTION 2020; 80:104197. [PMID: 31954917 DOI: 10.1016/j.meegid.2020.104197] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 01/12/2020] [Accepted: 01/15/2020] [Indexed: 02/07/2023]
Abstract
Antigenic diversity is a major concern in malaria vaccine development that requires to be considered in developing a malaria vaccine. Plasmodium falciparum thrombospondin-related adhesive protein (PfTRAP) is a leading malaria vaccine candidate antigen. In the current study, we investigated the level of genetic diversity and natural selection of pftrap sequences in P. falciparum isolates from Iran (n = 47). The gene diversity of Iranian pftrap sequences was also compared to available global pftrap sequences deposited in the GenBank or PlasmoDB databases (n = 220). Comparison of Iranian PfTRAP sequences with T9/96 reference sequence showed the presence of 35 amino acid changes in 32 positions and a limited variation in repeat sequences, leading to 13 distinct haplotypes. The overall nucleotide diversity (π) for the ectodomain of Iranian pftrap sequences was 0.00444 ± 0.00043, with the highest diversity in Domain IV. Alignment comparison of global PfTRAP sequences with T9/96 reference sequence indicated 96 amino acid replacements as well as extensive variable repeat sequences (9-23 repeats), which led to 192 haplotypes. Among the global isolates, the lowest nucleotide diversity was detected in French Guianan (0.00428 ± 0.00163) and Iranian (0.00444 ± 0.00043) pftrap sequences, and the most variation was observed in domains II and IV in all populations. The dN-dS value displayed the evidence of positive selection due to recombination and immune system pressure. The Fst analysis revealed a gene flow between African populations; however, genetic differentiation observed between Iranian and other populations probably was due to gene flow barriers. Both conserved and variable epitopes were predicted in B- and T-cell epitopes of PfTRAP antigen. The obtained results from this study could be helpful for developing a PfTRAP-based malaria vaccine.
Collapse
Affiliation(s)
- Akram Abouie Mehrizi
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, P.O.BOX 1316943551, Tehran, Iran.
| | - Azadeh Jafari Zadeh
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, P.O.BOX 1316943551, Tehran, Iran
| | - Sedigheh Zakeri
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, P.O.BOX 1316943551, Tehran, Iran
| | - Navid Dinparast Djadid
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, P.O.BOX 1316943551, Tehran, Iran
| |
Collapse
|
5
|
Crosby FL, Lundgren AM, Hoffman C, Pascual DW, Barbet AF. VirB10 vaccination for protection against Anaplasma phagocytophilum. BMC Microbiol 2018; 18:217. [PMID: 30563470 PMCID: PMC6299599 DOI: 10.1186/s12866-018-1346-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 11/16/2018] [Indexed: 12/18/2022] Open
Abstract
Background Human granulocytic anaplasmosis (HGA) is a tick-borne disease caused by the etiologic agent Anaplasma phagocytophilum. HGA was designated a nationally notifiable disease in the United States in 1998. Currently there are no vaccines available against HGA. Conserved membrane proteins that are subdominant in Anaplasma species, such as VirB9 and VirB10, may represent better vaccine targets than the variable immunodominant surface proteins. VirB9 and VirB10 are constituents of the Type 4 secretion system (T4SS) that is conserved amongst many intracellular bacteria and performs essential functions for invasion and survival in host cells. Results Immunogenicity and contribution to protection, provided after intramuscular vaccination of plasmid DNA encoding VirB9-1, VirB9-2, and VirB10 followed by inoculation of homologous recombinant proteins, in a prime-boost immunization strategy was evaluated in a murine model of HGA. Recombinant VirB9-1-, VirB9-2-, and VirB10-vaccinated mice developed antibody responses that specifically reacted with A. phagocytophilum organisms. However, only the mice vaccinated with VirB10 developed a significant increase in IFN-γ CD4+ T cells and partial protection against challenge with A. phagocytophilum. Conclusions This work provides evidence that A. phagocytophilum T4SS VirB10 is partially protective in a murine model against infection in an IFN-γ-dependent fashion and suggests that this protein may be a potential vaccine candidate against this and possibly other pathogenic bacteria with a T4SS.
Collapse
Affiliation(s)
- Francy L Crosby
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, 2015 SW 16th Avenue, Gainesville, FL, 32608, USA.
| | - Anna M Lundgren
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, 2015 SW 16th Avenue, Gainesville, FL, 32608, USA
| | - Carol Hoffman
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, 2015 SW 16th Avenue, Gainesville, FL, 32608, USA
| | - David W Pascual
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, 2015 SW 16th Avenue, Gainesville, FL, 32608, USA
| | - Anthony F Barbet
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, 2015 SW 16th Avenue, Gainesville, FL, 32608, USA
| |
Collapse
|
6
|
Volz A, Sutter G. Modified Vaccinia Virus Ankara: History, Value in Basic Research, and Current Perspectives for Vaccine Development. Adv Virus Res 2016; 97:187-243. [PMID: 28057259 PMCID: PMC7112317 DOI: 10.1016/bs.aivir.2016.07.001] [Citation(s) in RCA: 205] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Safety tested Modified Vaccinia virus Ankara (MVA) is licensed as third-generation vaccine against smallpox and serves as a potent vector system for development of new candidate vaccines against infectious diseases and cancer. Historically, MVA was developed by serial tissue culture passage in primary chicken cells of vaccinia virus strain Ankara, and clinically used to avoid the undesirable side effects of conventional smallpox vaccination. Adapted to growth in avian cells MVA lost the ability to replicate in mammalian hosts and lacks many of the genes orthopoxviruses use to conquer their host (cell) environment. As a biologically well-characterized mutant virus, MVA facilitates fundamental research to elucidate the functions of poxvirus host-interaction factors. As extremely safe viral vectors MVA vaccines have been found immunogenic and protective in various preclinical infection models. Multiple recombinant MVA currently undergo clinical testing for vaccination against human immunodeficiency viruses, Mycobacterium tuberculosis or Plasmodium falciparum. The versatility of the MVA vector vaccine platform is readily demonstrated by the swift development of experimental vaccines for immunization against emerging infections such as the Middle East Respiratory Syndrome. Recent advances include promising results from the clinical testing of recombinant MVA-producing antigens of highly pathogenic avian influenza virus H5N1 or Ebola virus. This review summarizes our current knowledge about MVA as a unique strain of vaccinia virus, and discusses the prospects of exploiting this virus as research tool in poxvirus biology or as safe viral vector vaccine to challenge existing and future bottlenecks in vaccinology.
Collapse
Affiliation(s)
- A Volz
- German Center for Infection Research (DZIF), Institute for Infectious Diseases and Zoonoses, LMU University of Munich, Munich, Germany
| | - G Sutter
- German Center for Infection Research (DZIF), Institute for Infectious Diseases and Zoonoses, LMU University of Munich, Munich, Germany.
| |
Collapse
|
7
|
Abstract
DNA plasmids can be used to induce a protective (or therapeutic) immune response by delivering genes encoding vaccine antigens. That naked DNA (without the refinement of coat proteins or host evasion systems) can cross from outside the cell into the nucleus and be expressed is particularly remarkable given the sophistication of the immune system in preventing infection by pathogens. As a result of the ease, low cost, and speed of custom gene synthesis, DNA vaccines dangle a tantalizing prospect of the next wave of vaccine technology, promising individual designer vaccines for cancer or mass vaccines with a rapid response time to emerging pandemics. There is considerable enthusiasm for the use of DNA vaccination as an approach, but this enthusiasm should be tempered by the successive failures in clinical trials to induce a potent immune response. The technology is evolving with the development of improved delivery systems that increase expression levels, particularly electroporation and the incorporation of genetically encoded adjuvants. This review will introduce some key concepts in the use of DNA plasmids as vaccines, including how the DNA enters the cell and is expressed, how it induces an immune response, and a summary of clinical trials with DNA vaccines. The review also explores the advances being made in vector design, delivery, formulation, and adjuvants to try to realize the promise of this technology for new vaccines. If the immunogenicity and expression barriers can be cracked, then DNA vaccines may offer a step change in mass vaccination.
Collapse
|
8
|
Frey SE, Wald A, Edupuganti S, Jackson LA, Stapleton JT, El Sahly H, El-Kamary SS, Edwards K, Keyserling H, Winokur P, Keitel W, Hill H, Goll JB, Anderson EL, Graham IL, Johnston C, Mulligan M, Rouphael N, Atmar R, Patel S, Chen W, Kotloff K, Creech CB, Chaplin P, Belshe RB. Comparison of lyophilized versus liquid modified vaccinia Ankara (MVA) formulations and subcutaneous versus intradermal routes of administration in healthy vaccinia-naïve subjects. Vaccine 2015; 33:5225-34. [PMID: 26143613 PMCID: PMC9533873 DOI: 10.1016/j.vaccine.2015.06.075] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Revised: 06/18/2015] [Accepted: 06/21/2015] [Indexed: 12/04/2022]
Abstract
Background Modified vaccinia Ankara (MVA) is being developed as a safer smallpox vaccine and is being placed in the US Strategic National Stockpile (SNS) as a liquid formulation for subcutaneous (SC) administration at a dose of 1 × 108 TCID50 in a volume of 0.5 mL. This study compared the safety and immunogenicity of the standard formulation, dose and route with both a more stable, lyophilized formulation and with an antigen-sparing intradermal (ID) route of administration. Methods 524 subjects were randomized to receive either a full dose of Lyophilized-SC, a full dose of Liquid-SC or 20% (2 × 107 TCID50 in 0.1 mL) of a full dose Liquid-ID MVA on Days 0 and 28. Safety and immunogenicity were followed through 180 days post second vaccination. Results Among the 3 groups, the proportion of subjects with moderate/severe functional local reactions was significantly different (P = 0.0013) between the Lyophilized-SC group (30.3%), the Liquid-SC group (13.8%) and Liquid-ID group (22.0%) only after first vaccination; and for moderate/severe measured erythema and/or induration after any vaccination (P = 0.0001) between the Lyophilized-SC group (58.2%), the Liquid-SC group (58.1%) and the Liquid-ID group (94.8%) and the reactions lasted longer in the Liquid-ID group. In the ID Group, 36.1% of subjects had mild injection site skin discoloration lasting ≥6 months. After second vaccination Day (42–208), geometric mean of peak neutralization titers were 87.8, 49.5 and 59.5 for the Lyophilized-SC, Liquid-SC and Liquid-ID groups, respectively, and the maximum number of responders based on peak titer in each group was 142/145 (97.9%), 142/149 (95.3%) and 138/146 (94.5%), respectively. At 180 days after the second vaccination, geometric mean neutralization titers declined to 11.7, 10.2 and 10.4 with only 54.3%, 39.2% and 35.2% of subjects remaining seropositive for the Lyophilized-SC, Liquid-SC and Liquid-ID groups, respectively. Both the Lyophilized-SC and Liquid-ID groups were considered non-inferior (primary objective) to the Liquid-SC group. Conclusions Transitioning to a lyophilized formulation, which has a longer shelf life, will not negatively impact immunogenicity. In a situation where insufficient vaccine is available, ID vaccination could be used, increasing the number of available doses of vaccine in the SNS 5-fold (i.e., from 20 million to 100 million doses).
Collapse
Affiliation(s)
- Sharon E Frey
- Saint Louis University School of Medicine, Department of Internal Medicine, St. Louis, MO, USA.
| | - Anna Wald
- University of Washington and Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Srilatha Edupuganti
- Emory University School of Medicine, Department of Medicine, Division of Infectious Diseases, Atlanta, GA, USA
| | | | - Jack T Stapleton
- University of Iowa and Iowa City VA Medical Center, Department of Internal Medicine, Iowa City, IA, USA
| | - Hana El Sahly
- Baylor College of Medicine, Departments of Molecular Virology and Microbiology and Medicine, Houston, TX, USA
| | - Samer S El-Kamary
- University of Maryland School of Medicine, Department of Epidemiology and Public Health, Center for Vaccine Development, Baltimore, MD, USA
| | - Kathryn Edwards
- Vanderbilt Vaccine Research Program, Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Harry Keyserling
- Emory University, Emory Children's Center, Department of Pediatrics, Atlanta, GA, USA
| | - Patricia Winokur
- University of Iowa and Iowa City VA Medical Center, Department of Internal Medicine, Iowa City, IA, USA
| | - Wendy Keitel
- Baylor College of Medicine, Departments of Molecular Virology and Microbiology and Medicine, Houston, TX, USA
| | | | | | - Edwin L Anderson
- Saint Louis University School of Medicine, Department of Internal Medicine, St. Louis, MO, USA
| | - Irene L Graham
- Saint Louis University School of Medicine, Department of Internal Medicine, St. Louis, MO, USA
| | - Christine Johnston
- University of Washington and Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Mark Mulligan
- Emory University School of Medicine, Department of Medicine, Division of Infectious Diseases, Atlanta, GA, USA
| | - Nadine Rouphael
- Emory University School of Medicine, Department of Medicine, Division of Infectious Diseases, Atlanta, GA, USA
| | - Robert Atmar
- Baylor College of Medicine, Departments of Molecular Virology and Microbiology and Medicine, Houston, TX, USA
| | - Shital Patel
- Baylor College of Medicine, Departments of Molecular Virology and Microbiology and Medicine, Houston, TX, USA
| | - Wilbur Chen
- University of Maryland School of Medicine, Center for Vaccine Development, Baltimore, MD, USA
| | - Karen Kotloff
- University of Maryland School of Medicine, Center for Vaccine Development, Baltimore, MD, USA
| | - C Buddy Creech
- Vanderbilt Vaccine Research Program, Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | | | - Robert B Belshe
- Saint Louis University School of Medicine, Department of Internal Medicine, St. Louis, MO, USA
| |
Collapse
|
9
|
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
|
10
|
Translating the immunogenicity of prime-boost immunization with ChAd63 and MVA ME-TRAP from malaria naive to malaria-endemic populations. Mol Ther 2014; 22:1992-2003. [PMID: 24930599 DOI: 10.1038/mt.2014.109] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2014] [Accepted: 06/08/2014] [Indexed: 01/17/2023] Open
Abstract
To induce a deployable level of efficacy, a successful malaria vaccine would likely benefit from both potent cellular and humoral immunity. These requirements are met by a heterologous prime-boost immunization strategy employing a chimpanzee adenovirus vector followed by modified vaccinia Ankara (MVA), both encoding the pre-erythrocytic malaria antigen ME-thrombospondin-related adhesive protein (TRAP), with high immunogenicity and significant efficacy in UK adults. We undertook two phase 1b open-label studies in adults in Kenya and The Gambia in areas of similar seasonal malaria transmission dynamics and have previously reported safety and basic immunogenicity data. We now report flow cytometry and additional interferon (IFN)-γ enzyme-linked immunospot (ELISPOT) data characterizing pre-existing and induced cellular immunity as well as anti-TRAP IgG responses. T-cell responses induced by vaccination averaged 1,254 spot-forming cells (SFC) per million peripheral blood mononuclear cells (PBMC) across both trials and flow cytometry revealed cytokine production from both CD4(+) and CD8(+) T cells with the frequency of CD8(+) IFN-γ-secreting monofunctional T cells (previously shown to associate with vaccine efficacy) particularly high in Kenyan adults. Immunization with ChAd63 and MVA ME-TRAP induced strong cellular and humoral immune responses in adults living in two malaria-endemic regions of Africa. This prime-boost approach targeting the pre-erythrocytic stage of the malaria life-cycle is now being assessed for efficacy in a target population.
Collapse
|
11
|
Moore AC, Hutchings CL. Combination vaccines: synergistic simultaneous induction of antibody and T-cell immunity. Expert Rev Vaccines 2014; 6:111-21. [PMID: 17280483 DOI: 10.1586/14760584.6.1.111] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Vaccines have traditionally been designed to induce antibody responses and have been licensed on their capacity to induce high titers of circulating antibody to the pathogen. With our increased knowledge of host-pathogen interactions, it became apparent that induction of the cellular arm of the immune response is crucial to the efficacy of vaccines against intracellular pathogens and for providing appropriate help for antibody induction. Diverging strategies emerged that concentrate on developing candidate vaccines that solely induce either cellular or humoral responses. As most microbes reside at some point in the infectious cycle in the extracellular as well as intracellular space, and there is interplay between antibody and T cells, it is now apparent that both arms of immunity are essential to effectively control and eliminate the infection. It is, therefore, necessary to develop vaccines that can effectively induce a broad adaptive immune response. For vaccines targeted at diseases of the developing world, such as HIV, tuberculosis and malaria, it is imperative that these vaccines are simple to deliver and cost effective, that is,that optimum T-cell and antibody immunity is achieved with the minimum number of vaccinations. Combination vaccines, where an antibody-inducing subunit protein vaccine is coadministered with a T-cell-inducing poxvirus-based vaccine fulfill these requirements and induce sterile immunity to pathogen challenge.
Collapse
Affiliation(s)
- Anne C Moore
- Wellcome Trust Centre for Human Genetics, Oxford University, Roosevelt Drive, Oxford OX2 7BN, UK.
| | | |
Collapse
|
12
|
Gómez CE, Perdiguero B, García-Arriaza J, Esteban M. Clinical applications of attenuated MVA poxvirus strain. Expert Rev Vaccines 2013; 12:1395-416. [PMID: 24168097 DOI: 10.1586/14760584.2013.845531] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The highly attenuated poxvirus strain modified vaccinia virus Ankara (MVA) has reached maturity as a vector delivery system and as a vaccine candidate against a broad spectrum of diseases. This has been largely recognized from research on virus-host cell interactions and immunological studies in pre-clinical and clinical trials. This review addresses the studies of MVA vectors used in phase I/II clinical trials, with the aim to provide the main findings obtained on their behavior when tested against relevant human diseases and cancer and also highlights the strategies currently implemented to improve the MVA immunogenicity. The authors assess that MVA vectors are progressing as strong vaccine candidates either alone or when administered in combination with other vectors.
Collapse
Affiliation(s)
- Carmen Elena Gómez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | | | | | | |
Collapse
|
13
|
Clinical development of Modified Vaccinia virus Ankara vaccines. Vaccine 2013; 31:4241-6. [PMID: 23523410 DOI: 10.1016/j.vaccine.2013.03.020] [Citation(s) in RCA: 116] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Accepted: 03/11/2013] [Indexed: 12/21/2022]
Abstract
The smallpox vaccine Vaccinia was successfully used to eradicate smallpox, but although very effective, it was a very reactogenic vaccine and responsible for the deaths of one or two people per million vaccinated. Modified Vaccinia virus Ankara (MVA) is a replication-deficient and attenuated derivative, also used in the smallpox eradication campaign and now being developed as a recombinant viral vector to produce vaccines against infectious diseases and cancer. Many clinical trials of these new vaccines have been conducted, and the findings of these trials are reviewed here. The safety of MVA is now well documented, immunogenicity is influenced by the dose and vaccination regimen, and information on the efficacy of MVA-vectored vaccines is now beginning to accumulate.
Collapse
|
14
|
Ogwang C, Afolabi M, Kimani D, Jagne YJ, Sheehy SH, Bliss CM, Duncan CJA, Collins KA, Garcia Knight MA, Kimani E, Anagnostou NA, Berrie E, Moyle S, Gilbert SC, Spencer AJ, Soipei P, Mueller J, Okebe J, Colloca S, Cortese R, Viebig NK, Roberts R, Gantlett K, Lawrie AM, Nicosia A, Imoukhuede EB, Bejon P, Urban BC, Flanagan KL, Ewer KJ, Chilengi R, Hill AVS, Bojang K. Safety and immunogenicity of heterologous prime-boost immunisation with Plasmodium falciparum malaria candidate vaccines, ChAd63 ME-TRAP and MVA ME-TRAP, in healthy Gambian and Kenyan adults. PLoS One 2013; 8:e57726. [PMID: 23526949 PMCID: PMC3602521 DOI: 10.1371/journal.pone.0057726] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2012] [Accepted: 01/24/2013] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Heterologous prime boost immunization with chimpanzee adenovirus 63 (ChAd63) and Modified vaccinia Virus Ankara (MVA) vectored vaccines is a strategy recently shown to be capable of inducing strong cell mediated responses against several antigens from the malaria parasite. ChAd63-MVA expressing the Plasmodium falciparum pre-erythrocytic antigen ME-TRAP (multiple epitope string with thrombospondin-related adhesion protein) is a leading malaria vaccine candidate, capable of inducing sterile protection in malaria naïve adults following controlled human malaria infection (CHMI). METHODOLOGY We conducted two Phase Ib dose escalation clinical trials assessing the safety and immunogenicity of ChAd63-MVA ME-TRAP in 46 healthy malaria exposed adults in two African countries with similar malaria transmission patterns. RESULTS ChAd63-MVA ME-TRAP was shown to be safe and immunogenic, inducing high-level T cell responses (median >1300 SFU/million PBMC). CONCLUSIONS ChAd63-MVA ME-TRAP is a safe and highly immunogenic vaccine regimen in adults with prior exposure to malaria. Further clinical trials to assess safety and immunogenicity in children and infants and protective efficacy in the field are now warranted. TRIAL REGISTRATION Pactr.org PACTR2010020001771828 Pactr.org PACTR201008000221638 ClinicalTrials.gov NCT01373879 NCT01373879 ClinicalTrials.gov NCT01379430 NCT01379430.
Collapse
Affiliation(s)
- Caroline Ogwang
- Kenya Medical Research Institute, Centre for Geographical Medical Research (Coast), Kilifi, Kenya
| | | | - Domtila Kimani
- Kenya Medical Research Institute, Centre for Geographical Medical Research (Coast), Kilifi, Kenya
| | | | - Susanne H. Sheehy
- Centre for Clinical Vaccinology and Tropical Medicine, The Jenner Institute, Churchill Hospital, Oxford, United Kingdom
- * E-mail:
| | - Carly M. Bliss
- The Jenner Institute Laboratories, University of Oxford, Old Road Campus Research Building, Oxford, United Kingdom
| | - Christopher J. A. Duncan
- Centre for Clinical Vaccinology and Tropical Medicine, The Jenner Institute, Churchill Hospital, Oxford, United Kingdom
| | - Katharine A. Collins
- The Jenner Institute Laboratories, University of Oxford, Old Road Campus Research Building, Oxford, United Kingdom
| | - Miguel A. Garcia Knight
- Kenya Medical Research Institute, Centre for Geographical Medical Research (Coast), Kilifi, Kenya
| | - Eva Kimani
- Kenya Medical Research Institute, Centre for Geographical Medical Research (Coast), Kilifi, Kenya
| | - Nicholas A. Anagnostou
- Centre for Clinical Vaccinology and Tropical Medicine, The Jenner Institute, Churchill Hospital, Oxford, United Kingdom
| | - Eleanor Berrie
- Clinical Biomanufacturing Facility, University of Oxford, Churchill Hospital, Oxford, United Kingdom
| | - Sarah Moyle
- Clinical Biomanufacturing Facility, University of Oxford, Churchill Hospital, Oxford, United Kingdom
| | - Sarah C. Gilbert
- The Jenner Institute Laboratories, University of Oxford, Old Road Campus Research Building, Oxford, United Kingdom
| | - Alexandra J. Spencer
- The Jenner Institute Laboratories, University of Oxford, Old Road Campus Research Building, Oxford, United Kingdom
| | - Peninah Soipei
- Kenya Medical Research Institute, Centre for Geographical Medical Research (Coast), Kilifi, Kenya
| | | | - Joseph Okebe
- Medical Research Council Unit, Fajara, The Gambia
| | | | | | | | - Rachel Roberts
- Centre for Clinical Vaccinology and Tropical Medicine, The Jenner Institute, Churchill Hospital, Oxford, United Kingdom
| | - Katherine Gantlett
- Centre for Clinical Vaccinology and Tropical Medicine, The Jenner Institute, Churchill Hospital, Oxford, United Kingdom
| | - Alison M. Lawrie
- Centre for Clinical Vaccinology and Tropical Medicine, The Jenner Institute, Churchill Hospital, Oxford, United Kingdom
| | - Alfredo Nicosia
- Okairòs AG, Rome, Italy
- CEINGE, Naples, Italy
- Department of Molecular Medicine and Medical Biotechnology, University Federico II Naples, Naples, Italy
| | | | - Philip Bejon
- Kenya Medical Research Institute, Centre for Geographical Medical Research (Coast), Kilifi, Kenya
| | - Britta C. Urban
- Kenya Medical Research Institute, Centre for Geographical Medical Research (Coast), Kilifi, Kenya
- Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | | | - Katie J. Ewer
- The Jenner Institute Laboratories, University of Oxford, Old Road Campus Research Building, Oxford, United Kingdom
| | - Roma Chilengi
- Kenya Medical Research Institute, Centre for Geographical Medical Research (Coast), Kilifi, Kenya
| | - Adrian V. S. Hill
- Centre for Clinical Vaccinology and Tropical Medicine, The Jenner Institute, Churchill Hospital, Oxford, United Kingdom
- The Jenner Institute Laboratories, University of Oxford, Old Road Campus Research Building, Oxford, United Kingdom
| | | |
Collapse
|
15
|
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
|
16
|
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]
|
17
|
Schwartz L, Brown GV, Genton B, Moorthy VS. A review of malaria vaccine clinical projects based on the WHO rainbow table. Malar J 2012; 11:11. [PMID: 22230255 PMCID: PMC3286401 DOI: 10.1186/1475-2875-11-11] [Citation(s) in RCA: 172] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2011] [Accepted: 01/09/2012] [Indexed: 12/14/2022] Open
Abstract
Development and Phase 3 testing of the most advanced malaria vaccine, RTS,S/AS01, indicates that malaria vaccine R&D is moving into a new phase. Field trials of several research malaria vaccines have also confirmed that it is possible to impact the host-parasite relationship through vaccine-induced immune responses to multiple antigenic targets using different platforms. Other approaches have been appropriately tested but turned out to be disappointing after clinical evaluation. As the malaria community considers the potential role of a first-generation malaria vaccine in malaria control efforts, it is an apposite time to carefully document terminated and ongoing malaria vaccine research projects so that lessons learned can be applied to increase the chances of success for second-generation malaria vaccines over the next 10 years. The most comprehensive resource of malaria vaccine projects is a spreadsheet compiled by WHO thanks to the input from funding agencies, sponsors and investigators worldwide. This spreadsheet, available from WHO's website, is known as "the rainbow table". By summarizing the published and some unpublished information available for each project on the rainbow table, the most comprehensive review of malaria vaccine projects to be published in the last several years is provided below.
Collapse
Affiliation(s)
- Lauren Schwartz
- Initiative for Vaccine Research, Department of Immunization, Vaccines & Biologicals, World Health Organization, Avenue Appia 20, 1211-CH 27, Geneva, Switzerland
| | | | | | | |
Collapse
|
18
|
Rowland R, Brittain N, Poulton ID, Minassian AM, Sander C, Porter DW, Williams N, Satti I, Pathan AA, Lawrie AM, McShane H. A review of the tolerability of the candidate TB vaccine, MVA85A compared with BCG and Yellow Fever vaccines, and correlation between MVA85A vaccine reactogenicity and cellular immunogenicity. ACTA ACUST UNITED AC 2012. [DOI: 10.1016/j.trivac.2012.07.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
19
|
Almeida APMM, Bruna-Romero O. Synergism/complementarity of recombinant adenoviral vectors and other vaccination platforms during induction of protective immunity against malaria. Mem Inst Oswaldo Cruz 2011; 106 Suppl 1:193-201. [PMID: 21881774 DOI: 10.1590/s0074-02762011000900024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2011] [Accepted: 06/15/2011] [Indexed: 12/19/2022] Open
Abstract
The lack of immunogenicity of most malaria antigens and the complex immune responses required for achieving protective immunity against this infectious disease have traditionally hampered the development of an efficient human malaria vaccine. The current boom in development of recombinant viral vectors and their use in prime-boost protocols that result in enhanced immune outcomes have increased the number of malaria vaccine candidates that access pre-clinical and clinical trials. In the frontline, adenoviruses and poxviruses seem to be giving the best immunization results in experimental animals and their mutual combination, or their combination with recombinant proteins (formulated in adjuvants and given in sequence or being given as protein/virus admixtures), has been shown to reach unprecedented levels of anti-malaria immunity that predictably will be somehow reproduced in the human setting. However, all this optimism was previously seen in the malaria vaccine development field without many real applicable results to date. We describe here the current state-of-the-art in the field of recombinant adenovirus research for malaria vaccine development, in particular referring to their use in combination with other immunogens in heterologous prime-boost protocols, while trying to simultaneously show our contributions and point of view on this subject.
Collapse
|
20
|
Porter DW, Thompson FM, Berthoud TK, Hutchings CL, Andrews L, Biswas S, Poulton I, Prieur E, Correa S, Rowland R, Lang T, Williams J, Gilbert SC, Sinden RE, Todryk S, Hill AVS. A human Phase I/IIa malaria challenge trial of a polyprotein malaria vaccine. Vaccine 2011; 29:7514-22. [PMID: 21501642 PMCID: PMC3195259 DOI: 10.1016/j.vaccine.2011.03.083] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2010] [Revised: 02/12/2011] [Accepted: 03/22/2011] [Indexed: 11/30/2022]
Abstract
We examined the safety, immunogenicity and efficacy of a prime-boost vaccination regime involving two poxvirus malaria subunit vaccines, FP9-PP and MVA-PP, expressing the same polyprotein consisting of six pre-erythrocytic antigens from Plasmodium falciparum. Following safety assessment of single doses, 15 volunteers received a heterologous prime-boost vaccination regime and underwent malaria sporozoite challenge. The vaccines were safe but interferon-γ ELISPOT responses were low compared to other poxvirus vectors, despite targeting multiple antigens. There was no vaccine efficacy as measured by delay in time to parasitaemia. A number of possible explanations are discussed, including the very large insert size of the polyprotein transgene.
Collapse
Affiliation(s)
- David W Porter
- Centre for Clinical Vaccinology and Tropical Medicine, Churchill Hospital, Old Road, Oxford, OX3 7LJ, UK.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Cavenaugh JS, Awi D, Mendy M, Hill AVS, Whittle H, McConkey SJ. Partially randomized, non-blinded trial of DNA and MVA therapeutic vaccines based on hepatitis B virus surface protein for chronic HBV infection. PLoS One 2011; 6:e14626. [PMID: 21347224 PMCID: PMC3039644 DOI: 10.1371/journal.pone.0014626] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2008] [Accepted: 11/25/2010] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Chronic HBV infects 350 million people causing cancer and liver failure. We aimed to assess the safety and efficacy of plasmid DNA (pSG2.HBs) vaccine, followed by recombinant modified vaccinia virus Ankara (MVA.HBs), encoding the surface antigen of HBV as therapy for chronic HBV. A secondary goal was to characterize the immune responses. METHODS Firstly 32 HBV e antigen negative (eAg(-)) participants were randomly assigned to one of four groups: to receive vaccines alone, lamivudine (3TC) alone, both, or neither. Later 16 eAg(+) volunteers in two groups received either 3TC alone or both 3TC and vaccines. Finally, 12 eAg(-) and 12 eAg(+) subjects were enrolled into higher-dose treatment groups. Healthy but chronically HBV-infected males between the ages of 15-25 who lived in the western part of The Gambia were eligible. Participants in some groups received 1 mg or 2 mg of pSG2.HBs intramuscularly twice followed by 5×10(7) pfu or 1.5×10(8) pfu of MVA.HBs intradermally at 3-weekly intervals with or without concomitant 3TC for 11-14 weeks. Intradermal rabies vaccine was administered to a negative control group. Safety was assessed clinically and biochemically. The primary measure of efficacy was a quantitative PCR assay of plasma HBV. Immunity was assessed by IFN-γ ELISpot and intracellular cytokine staining. RESULTS Mild local and systemic adverse events were observed following the vaccines. A small shiny scar was observed in some cases after MVA.HBs. There were no significant changes in AST or ALT. HBeAg was lost in one participant in the higher-dose group. As expected, the 3TC therapy reduced viraemia levels during therapy, but the prime-boost vaccine regimen did not reduce the viraemia. The immune responses were variable. The majority of IFN-γ was made by antigen non-specific CD16(+) cells (both CD3(+) and CD3(-)). CONCLUSIONS The vaccines were well tolerated but did not control HBV infection. TRIAL REGISTRATION ISRCTN ISRCTN67270384.
Collapse
Affiliation(s)
- James S. Cavenaugh
- Medical Research Council Laboratories, Banjul,
The Gambia
- Centre for Clinical Vaccinology and Tropical
Medicine, University of Oxford, Oxford, United Kingdom
- David H. Smith Center for Vaccine Biology and
Immunology, School of Medicine and Dentistry, University of Rochester,
Rochester, New York, United States of America
| | - Dorka Awi
- Medical Research Council Laboratories, Banjul,
The Gambia
- Institute of Maternal and Child Health,
University of Port Harcourt, Post Harcourt, Nigeria
| | - Maimuna Mendy
- Medical Research Council Laboratories, Banjul,
The Gambia
- International Agency for Research on Cancer,
Lyon, France
| | - Adrian V. S. Hill
- Centre for Clinical Vaccinology and Tropical
Medicine, University of Oxford, Oxford, United Kingdom
- The Wellcome Trust Centre for Human Genetics,
Oxford, United Kingdom
| | - Hilton Whittle
- Medical Research Council Laboratories, Banjul,
The Gambia
| | - Samuel J. McConkey
- Medical Research Council Laboratories, Banjul,
The Gambia
- Centre for Clinical Vaccinology and Tropical
Medicine, University of Oxford, Oxford, United Kingdom
- Department of International Health and
Tropical Medicine, Royal College of Surgeons in Ireland, Dublin,
Ireland
| |
Collapse
|
22
|
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
|
23
|
Guilbride DL, Gawlinski P, Guilbride PDL. Why functional pre-erythrocytic and bloodstage malaria vaccines fail: a meta-analysis of fully protective immunizations and novel immunological model. PLoS One 2010; 5:e10685. [PMID: 20502667 PMCID: PMC2873430 DOI: 10.1371/journal.pone.0010685] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2009] [Accepted: 04/16/2010] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Clinically protective malaria vaccines consistently fail to protect adults and children in endemic settings, and at best only partially protect infants. METHODOLOGY/PRINCIPAL FINDINGS We identify and evaluate 1916 immunization studies between 1965-February 2010, and exclude partially or nonprotective results to find 177 completely protective immunization experiments. Detailed reexamination reveals an unexpectedly mundane basis for selective vaccine failure: live malaria parasites in the skin inhibit vaccine function. We next show published molecular and cellular data support a testable, novel model where parasite-host interactions in the skin induce malaria-specific regulatory T cells, and subvert early antigen-specific immunity to parasite-specific immunotolerance. This ensures infection and tolerance to reinfection. Exposure to Plasmodium-infected mosquito bites therefore systematically triggers immunosuppression of endemic vaccine-elicited responses. The extensive vaccine trial data solidly substantiate this model experimentally. CONCLUSIONS/SIGNIFICANCE We conclude skinstage-initiated immunosuppression, unassociated with bloodstage parasites, systematically blocks vaccine function in the field. Our model exposes novel molecular and procedural strategies to significantly and quickly increase protective efficacy in both pipeline and currently ineffective malaria vaccines, and forces fundamental reassessment of central precepts determining vaccine development. This has major implications for accelerated local eliminations of malaria, and significantly increases potential for eradication.
Collapse
|
24
|
Singhal N, Bisht D, Joshi B. Immunoprophylaxis of tuberculosis: an update of emerging trends. Arch Immunol Ther Exp (Warsz) 2010; 58:97-106. [PMID: 20140756 DOI: 10.1007/s00005-010-0068-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2009] [Accepted: 07/06/2009] [Indexed: 11/25/2022]
Abstract
Developing effective prophylactics to combat tuberculosis is currently in an exploratory stage. The HIV pandemic and emergence of multi- and extensively drug-resistant strains of Mycobacterium tuberculosis indicate that the current preventive measures against this ever-evolving pathogen are inadequate. The currently available vaccine BCG in its present form affords variable protection which usually wanes with aging. Various reasons have been cited to explain the discrepancies in the efficacy of BCG, including generic differences in the different BCG vaccine strains used in immunization program throughout the world. The low efficacy of BCG vaccine has promoted the search for novel vaccines for tuberculosis. The search strategies aim at completely replacing the existing vaccine and/or augmenting/improving the current BCG vaccine. Among new vaccine candidates are live attenuated M. tuberculosis vaccines, recombinant BCG, DNA vaccines, subunit vaccine, and fusion protein-based vaccines. More than 200 new vaccine candidates have been developed as a result of research work over the past few years. To date, at least eight vaccine candidates are undergoing clinical evaluation, with a few of them successfully qualifying in the first phase of clinical testing. These recent advances present an optimistic insight whereby a new tuberculosis vaccine might be expected to be available for public use in the next few years.
Collapse
Affiliation(s)
- Neelja Singhal
- Department of Biochemistry, National JALMA Institute for Leprosy and Other Mycobacterial Diseases, Indian Council of Medical Research, Tajganj, Agra 282001, India
| | | | | |
Collapse
|
25
|
Pandey A, Singh N, Sambhara S, Mittal SK. Egg-independent vaccine strategies for highly pathogenic H5N1 influenza viruses. HUMAN VACCINES 2010; 6:178-88. [PMID: 19875936 PMCID: PMC2888842 DOI: 10.4161/hv.6.2.9899] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The emergence of a highly pathogenic H5N1 influenza virus in Hong Kong in 1997 and the subsequent appearance of other H5N1 strains and their spread to several countries in southeast Asia, Africa, the Middle East and Europe has evoked fear of a global influenza pandemic. Vaccines offer the best hope to combat the threat of an influenza pandemic. However, the global demand for a pandemic vaccine cannot be fulfilled by the current egg-based vaccine manufacturing strategies, thus creating a need to explore alternative technologies for vaccine production and delivery. Several egg-independent vaccine approaches such as cell culture-derived whole virus or subvirion vaccines, recombinant protein-based vaccines, virus-like particle (VLP) vaccines, DNA vaccines and viral vector-based vaccines are currently being investigated and appear promising both in preclinical and clinical studies. The present review will highlight the various egg-independent alternative vaccine approaches for pandemic influenza.
Collapse
Affiliation(s)
| | | | | | - Suresh K. Mittal
- Correspondence: Suresh K. Mittal, Department of Comparative Pathobiology, School of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA, Tel: 765-496-2894, Fax: 765-494-9830, , Suryaprakash Sambhara, Influenza Division, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA, Tel: 404-639-3800, Fax: 404-639-5180,
| |
Collapse
|
26
|
Abstract
Traditional vaccine technologies have resulted in an impressive array of efficacious vaccines against a variety of infectious agents. However, several potentially deadly pathogens, including retroviruses and parasites, have proven less amenable to the application of traditional vaccine platforms, indicating the need for new approaches. Viral vectors represent an attractive way to deliver and present vaccine antigens that may offer advantages over traditional platforms. Due to their ability to induce strong cell-mediated immunity (CMI) in addition to antibodies, viral vectors may be suitable for infectious agents, such as malaria parasites, where potent CMI is required for protection. Poxvirus-vectored malaria vaccines have been the most extensively studied in the clinic, achieving significant reductions in liver-stage parasite burden. More recently, adenovirus-vectored malaria vaccines have entered clinical testing. The most promising approach - heterologous prime-boost regimens, in which different viral vectors are sequentially paired with each other or with DNA or recombinant protein vaccines - is now being explored, and could provide high-grade protection, if findings in animal models are translatable to humans. Significant barriers remain, however, such as pre-existing immunity to the vector particle and an unexplained safety signal observed in one trial suggesting an increased risk of HIV acquisition in volunteers with pre-existing immunity to the vector.
Collapse
Affiliation(s)
- K J Limbach
- U.S. Military Malaria Vaccine Program, Naval Medical Research Center, 503 Robert Grant Avenue, Silver Spring, MD 20910-7500, USA.
| | | |
Collapse
|
27
|
Roshorm Y, Hong JP, Kobayashi N, McMichael AJ, Volsky DJ, Potash MJ, Takiguchi M, Hanke T. Novel HIV-1 clade B candidate vaccines designed for HLA-B*5101(+) patients protected mice against chimaeric ecotropic HIV-1 challenge. Eur J Immunol 2009; 39:1831-40. [PMID: 19585509 DOI: 10.1002/eji.200939309] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Novel candidate HIV-1 vaccines have been constructed, which are tailor-designed for HLA-B*5101(+) patients infected with HIV-1 clade B. These vaccines employ novel immunogen HIVB-B*5101 derived from consensus HIV-1 clade B Gag p17 and p24 regions coupled to two Pol-derived B*5101-restricted epitopes, which are together with a third B*5101 epitope in Gag dominant in HIV-1-infected long-term non-progressing patients. Both plasmid DNA and modified vaccinia virus Ankara (MVA) vectors supported high expression levels of the HIVB-B*5101 immunogen in cultured cells. Heterologous DNA prime-recombinant MVA boost regimen induced efficiently HIV-1-specific CD8(+) T-cell responses in BALB/c mice. These vaccine-elicited T cells were multifunctional, killed efficiently target cells in vivo, and protected mice against challenge with ecotropic HIV-1/NL4-3 and ecotropic HIV-1/NDK chimaeric viruses with HIV-1 clade B or D backbones, respectively, and ecotropic murine leukemia virus gp80 envelope, and therefore did so in the absence of anti-HIV-1 gp120 antibodies. These results support further development of HIVB-B*5101 vaccines in combined heterologous-modality regimens. The use of allele-specific vaccines in humans is discussed in the context of other developments in the HIV-1 field.
Collapse
Affiliation(s)
- Yaowaluck Roshorm
- Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe, UK
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Winstone N, Guimarães-Walker A, Roberts J, Brown D, Loach V, Goonetilleke N, Hanke T, McMichael AJ. Increased detection of proliferating, polyfunctional, HIV-1-specific T cells in DNA-modified vaccinia virus Ankara-vaccinated human volunteers by cultured IFN-γ ELISPOT assay. Eur J Immunol 2009; 39:975-85. [DOI: 10.1002/eji.200839167] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
29
|
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
|
30
|
A heterologous DNA prime/protein boost immunization strategy for rhesus cytomegalovirus. Vaccine 2008; 26:6013-25. [PMID: 18760319 DOI: 10.1016/j.vaccine.2008.07.103] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2008] [Revised: 07/29/2008] [Accepted: 07/30/2008] [Indexed: 12/11/2022]
Abstract
A previous study in nonhuman primates demonstrated that genetic immunization against the rhesus cytomegalovirus phosphoprotein 65-2 (pp65-2) and glycoprotein B (gB) antigens both stimulated antigen-specific antibodies and CD8 T cell responses, and significantly reduced plasma viral loads following intravenous challenge with RhCMV. It was also noted in this study that weak CD4 T cell and neutralizing antibody responses were generated by DNA alone. To broaden the type of immune responses, a DNA prime/protein boost strategy was used in seronegative macaques, consisting of four DNA immunizations against pp65-2, gB, and immediate-early 1 (IE1), followed by two boosts with formalin-inactivated RhCMV virions. This heterologous prime/boost strategy elicited robust antigen-specific CD4 and CD8 T cell responses in addition to biologically relevant neutralizing antibody titers. Animals were challenged with RhCMV delivered into four sites via a subcutaneous route. Skin biopsies of one of the inoculation sites 7 days post challenge revealed marked differences in the level of RhCMV replication between the vaccinated and control monkeys. Whereas the inoculation site in the controls was noted for a prominent inflammatory response and numerous cytomegalic, antigen-positive (IE1) cells, the inoculation site in the vaccinees was characterized by an absence of inflammation and antigen-positive cells. All five vaccinees developed robust recall responses to viral antigens, and four of them exhibited long-term viral immune responses consistent with effective control of viral expression and replication. These results demonstrate that a heterologous DNA prime/protein boost strategy greatly expands the breadth of antiviral immune responses and greatly reduces the level of viral replication at the primary site of challenge infection.
Collapse
|
31
|
Rosada RS, de la Torre LG, Frantz FG, Trombone APF, Zárate-Bladés CR, Fonseca DM, Souza PRM, Brandão IT, Masson AP, Soares EG, Ramos SG, Faccioli LH, Silva CL, Santana MHA, Coelho-Castelo AAM. Protection against tuberculosis by a single intranasal administration of DNA-hsp65 vaccine complexed with cationic liposomes. BMC Immunol 2008; 9:38. [PMID: 18647414 PMCID: PMC2500095 DOI: 10.1186/1471-2172-9-38] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2008] [Accepted: 07/22/2008] [Indexed: 02/19/2023] Open
Abstract
Background The greatest challenges in vaccine development include optimization of DNA vaccines for use in humans, creation of effective single-dose vaccines, development of delivery systems that do not involve live viruses, and the identification of effective new adjuvants. Herein, we describe a novel, simple technique for efficiently vaccinating mice against tuberculosis (TB). Our technique consists of a single-dose, genetic vaccine formulation of DNA-hsp65 complexed with cationic liposomes and administered intranasally. Results We developed a novel and non-toxic formulation of cationic liposomes, in which the DNA-hsp65 vaccine was entrapped (ENTR-hsp65) or complexed (COMP-hsp65), and used to immunize mice by intramuscular or intranasal routes. Although both liposome formulations induced a typical Th1 pattern of immune response, the intramuscular route of delivery did not reduce the number of bacilli. However, a single intranasal immunization with COMP-hsp65, carrying as few as 25 μg of plasmid DNA, leads to a remarkable reduction of the amount of bacilli in lungs. These effects were accompanied by increasing levels of IFN-γ and lung parenchyma preservation, results similar to those found in mice vaccinated intramuscularly four times with naked DNA-hsp65 (total of 400 μg). Conclusion Our objective was to overcome the significant obstacles currently facing DNA vaccine development. Our results in the mouse TB model showed that a single intranasal dose of COMP-hsp65 elicited a cellular immune response that was as strong as that induced by four intramuscular doses of naked-DNA. This formulation allowed a 16-fold reduction in the amount of DNA administered. Moreover, we demonstrated that this vaccine is safe, biocompatible, stable, and easily manufactured at a low cost. We believe that this strategy can be applied to human vaccines to TB in a single dose or in prime-boost protocols, leading to a tremendous impact on the control of this infectious disease.
Collapse
Affiliation(s)
- Rogério S Rosada
- Núcleo de Pesquisas em Tuberculose, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, São Paulo,
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Wang Z, Zhou W, Srivastava T, La Rosa C, Mandarino A, Forman SJ, Zaia JA, Britt WJ, Diamond DJ. A fusion protein of HCMV IE1 exon4 and IE2 exon5 stimulates potent cellular immunity in an MVA vaccine vector. Virology 2008; 377:379-90. [PMID: 18538366 DOI: 10.1016/j.virol.2008.04.034] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2008] [Revised: 03/25/2008] [Accepted: 04/24/2008] [Indexed: 11/25/2022]
Abstract
A therapeutic CMV vaccine incorporating an antigenic repertoire capable of eliciting a cellular immune response has yet to be successfully implemented for patients who already have acquired an infection. To address this problem, we have developed a vaccine candidate derived from modified vaccinia Ankara (MVA) that expresses three immunodominant antigens (pp65, IE1, IE2) from CMV. The novelty of this vaccine is the fusion of two adjacent exons from the immediate-early region of CMV, their successful expression in MVA, and robust immunogenicity in both primary and memory response models. Evaluation of the immunogenicity of the viral vaccine in mouse models shows that it can stimulate primary immunity against all three antigens in both the CD4(+) and CD8(+) T cell subsets. Evaluation of human PBMC from healthy CMV-positive donors or patients within 6 months of receiving hematopoietic cell transplant shows robust stimulation of existing CMV-specific CD4(+) and CD8(+) T cell subsets.
Collapse
Affiliation(s)
- Z Wang
- Laboratory of Vaccine Research, Division of Virology, Beckman Research Institute of the City of Hope, California 91010, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Lu S, Wang S, Grimes-Serrano JM. Current progress of DNA vaccine studies in humans. Expert Rev Vaccines 2008; 7:175-91. [PMID: 18324888 DOI: 10.1586/14760584.7.2.175] [Citation(s) in RCA: 114] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Despite remarkable progress in the field of DNA vaccine research since its discovery in the early 1990 s, the formal acceptance of this novel technology as a new modality of human vaccines depends on the successful demonstration of its safety and efficacy in advanced clinical trials. Although clinical trials conducted so far have provided overwhelming evidence that DNA vaccines are well tolerated and have an excellent safety profile, the early designs of DNA vaccines failed to demonstrate sufficient immunogenicity in humans. However, studies conducted over the last few years have led to promising results, particularly when DNA vaccines were used in combination with other forms of vaccines. Here, we provide a review of the data from reported DNA vaccine clinical studies with an emphasis on the ability of DNA vaccines to elicit antigen-specific, cell-mediated and antibody responses in humans. The majority of these trials are designed to test candidate vaccines against several major human pathogens and the remaining studies tested the immunogenicity of therapeutic vaccines against cancer.
Collapse
Affiliation(s)
- Shan Lu
- Department of Medicine, University of Massachusetts Medical School, 364 Plantation Street, LRB 304, Worcester, MA 01605, USA.
| | | | | |
Collapse
|
34
|
Hafalla JCR, Rai U, Bernal-Rubio D, Rodriguez A, Zavala F. Efficient development of plasmodium liver stage-specific memory CD8+ T cells during the course of blood-stage malarial infection. J Infect Dis 2008; 196:1827-35. [PMID: 18190264 DOI: 10.1086/522965] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Immunity to Plasmodium liver stages in individuals in malaria-endemic areas is inextricably linked to concomitant blood-stage parasitemia. Although Plasmodium sporozoite infection induces measurable CD8+ T cell responses, the development of memory T cells during active erythrocytic infection remains uncharacterized. Using transgenic T cells, we assessed antigen-specific effector CD8+ T cell responses induced by normal (NorSpz) and radiation-attenuated (IrrSpz) Plasmodium yoelii sporozoites. The magnitude, phenotypic activation, and differentiation pathway of CD8+ T cells were similarly induced by NorSpz and IrrSpz. Moreover, in normal mice, memory T cells elicited after priming with NorSpz and IrrSpz generated identical recall responses after a heterologous boost strategy. Furthermore, these recall responses exhibited comparable in vivo antiparasite activity. Our results indicate that sporozoites that retain their infective capacity induce memory CD8+ T cells that are robustly recalled by secondary immunization. Thus, erythrocytic infection does not preclude the establishment of memory CD8+ T cell responses to malarial liver stages.
Collapse
Affiliation(s)
- Julius C R Hafalla
- Department of Medical Parasitology, New York University School of Medicine, 341 E. 25th Street, New York, NY 10010, USA
| | | | | | | | | |
Collapse
|
35
|
Dubovsky F, Malkin E. Malaria vaccines. Vaccines (Basel) 2008. [DOI: 10.1016/b978-1-4160-3611-1.50056-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
36
|
Przybylowski M, Bartido S, Borquez-Ojeda O, Sadelain M, Rivière I. Production of clinical-grade plasmid DNA for human Phase I clinical trials and large animal clinical studies. Vaccine 2007; 25:5013-24. [PMID: 17537555 DOI: 10.1016/j.vaccine.2007.04.077] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2006] [Revised: 04/16/2007] [Accepted: 04/18/2007] [Indexed: 11/30/2022]
Abstract
The use of plasmid DNA as vaccines for the treatment of cancer and infectious diseases is on the rise. In order to facilitate the manufacture of clinical-grade plasmid DNA for Phase I clinical trials, we developed a process whereby >200 mg plasmid could be produced in a single production run under Good Manufacturing Practices. A dedicated cleanroom (Class 10,000 with Class 100 biosafety cabinet) is utilized for production of the bacterial cell bank, fermentation, harvest/lysis of the biomass, and downstream purification. Fermentation requires three 16-18 h runs (approximately 12 L each) in shaker-flasks, yielding approximately 60 g bacterial paste following batch centrifugation. The biomass is alkaline-lysed, pooled, and the resulting flocculent precipitate is separated by a novel vacuum step, followed by depth-filtration. Downstream processing includes anion-exchange chromatography, utilizing Qiagen silica-based resin, and precipitation with isopropanol. Following precipitation, the DNA is harvested by centrifugation, dried, formulated, and sterile-filtered using a Sartorius Sartobran 150 filter prior to Final-Filling. All processing steps utilize sterilized, single-use components. This process results in a product manufactured according to regulatory guidelines. The plasmid DNA is sterile with >or=95% supercoiled DNA, an A260/A280 ratio>or=1.9, undetectable or extremely low residual endotoxin, RNA, genomic DNA, protein, and antibiotic. Residual solvent levels are negligible. The product yields the predicted profile upon restriction-enzyme digestion, is biologically active upon transfection and remains stable for several years at -20 degrees C. We have therefore developed a reproducible and cost effective process to manufacture clinical-grade plasmid DNA. This process can be adapted by other academic centers for human or large animal clinical trials.
Collapse
Affiliation(s)
- Mark Przybylowski
- Gene Transfer and Somatic Cell Engineering Facility, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY 10021, USA
| | | | | | | | | |
Collapse
|
37
|
El-Gogo S, Staib C, Meyr M, Erfle V, Sutter G, Adler H. Recombinant murine gammaherpesvirus 68 (MHV-68) as challenge virus to test efficacy of vaccination against chronic virus infections in the mouse model. Vaccine 2007; 25:3934-45. [PMID: 17433507 DOI: 10.1016/j.vaccine.2007.02.054] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2006] [Revised: 01/19/2007] [Accepted: 02/20/2007] [Indexed: 11/18/2022]
Abstract
Efficient vaccines against AIDS, Hepatitis C and other persistent virus infections are urgently needed. Vaccine development has been especially hampered by the lack of suitable small animal models to reliably test the protective capacity of candidate vaccines against such chronic viral infections. A natural mouse pathogen such as MHV-68 that persists lifelong after infection, appears to be a particularly promising candidate for a more relevant model system. Here, we investigated infections with recombinant MHV-68 as novel mouse challenge model to test the efficacy of heterologous vaccines based on recombinant modified vaccinia virus Ankara (MVA). To apply ovalbumin (OVA) as a model antigen, we constructed the recombinant virus MHV-68-OVA by BAC technology and characterized genetic stability and replicative capacity of the virus in vitro and in vivo. We demonstrated the ability of MHV-68-OVA to produce ovalbumin upon tissue culture infection. Moreover, the use of MHV-68-OVA-infected target cells allowed for efficient ex vivo amplification of OVA-specific, MHC class I-restricted CD8 T cells derived from MVA-OVA-vaccinated C57BL/6 mice. Finally, we immunized C57BL/6 mice with MVA-OVA and challenged the animals with MHV-68-OVA testing different time points and routes of infection. Vaccinated mice were infected with MHV-68-OVA but showed reduced viral loads in the acute and latent phase of challenge infection. These data strongly suggest the usefulness of the MHV-68 challenge model for further evaluation of recombinant vaccines against persisting virus infections.
Collapse
Affiliation(s)
- Susanne El-Gogo
- Institute of Virology, Technical University München, Germany
| | | | | | | | | | | |
Collapse
|
38
|
Hanke T, McMichael AJ, Dorrell L. Clinical experience with plasmid DNA- and modified vaccinia virus Ankara-vectored human immunodeficiency virus type 1 clade A vaccine focusing on T-cell induction. J Gen Virol 2007; 88:1-12. [PMID: 17170430 DOI: 10.1099/vir.0.82493-0] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Candidate human immunodeficiency virus type 1 (HIV-1) vaccines focusing on T-cell induction, constructed as pTHr.HIVA DNA and modified vaccinia virus Ankara (MVA).HIVA, were delivered in a heterologous prime-boost regimen. The vaccines were tested in several hundred healthy or HIV-1-infected volunteers in Europe and Africa. Whilst larger trials of hundreds of volunteers suggested induction of HIV-1-specific T-cell responses in <15 % of healthy vaccinees, a series of small, rapid trials in 12-24 volunteers at a time with a more in-depth analysis of vaccine-elicited T-cell responses proved to be highly informative and provided more encouraging results. These trials demonstrated that the pTHr.HIVA vaccine alone primed consistently weak and mainly CD4(+), but also CD8(+) T-cell responses, and the MVA.HIVA vaccine delivered a consistent boost to both CD4(+) and CD8(+) T cells, which was particularly strong in HIV-1-infected patients. Thus, whilst the search is on for ways to enhance T-cell priming, MVA is a useful boosting vector for human subunit genetic vaccines.
Collapse
Affiliation(s)
- Tomáš Hanke
- Weatherall Institute of Molecular Medicine, MRC Human Immunology Unit, University of Oxford, The John Radcliffe, Oxford OX3 9DS, UK
| | - Andrew J McMichael
- Weatherall Institute of Molecular Medicine, MRC Human Immunology Unit, University of Oxford, The John Radcliffe, Oxford OX3 9DS, UK
| | - Lucy Dorrell
- Weatherall Institute of Molecular Medicine, MRC Human Immunology Unit, University of Oxford, The John Radcliffe, Oxford OX3 9DS, UK
| |
Collapse
|
39
|
Abstract
BACKGROUND Vaccines against all stages of the malaria parasite are in development, mainly for Plasmodium falciparum, which causes the most serious form of malaria. Pre-erythrocytic vaccines act to prevent or delay a malaria attack by attacking the sporozoite and liver stages before the parasite reaches the bloodstream. OBJECTIVES To assess the efficacy and safety of pre-erythrocytic malaria vaccines against any type of human malaria. SEARCH STRATEGY In March 2006, we searched the Cochrane Infectious Diseases Group Specialized Register, CENTRAL (The Cochrane Library 2006, Issue 1), MEDLINE, EMBASE, LILACS, and the Science Citation Index. We also searched conference proceedings and reference lists of articles, and contacted organizations and researchers in the field. SELECTION CRITERIA Randomized controlled trials comparing pre-erythrocytic vaccines with placebo, control vaccine, or routine antimalarial control measures in people of any age receiving an artificial challenge or natural exposure to malaria infection. DATA COLLECTION AND ANALYSIS Both authors independently assessed trial quality and extracted data. Results of meta-analyses were expressed as relative risks with 95% confidence intervals (CI) using an intention-to-treat analysis. MAIN RESULTS Nine safety and efficacy trials, and two safety trials, with over 3000 participants were included. In semi-immune children, RTS,S vaccine reduced clinical episodes of malaria by 26% (95% CI 13% to 37%) and severe malaria by 58% (95% CI 15% to 79%) for up to 18 months. Prevalence of parasitaemia was also reduced by 26% (95% CI 11% to 38%) at six months after immunization. RTS,S also reduced clinical malaria episodes by 63% (95% CI 18% to 83%) in semi-immune adult men in the second year of follow up after a booster dose. No severe adverse events were judged to be related to RTS,S vaccine, although the frequencies of injection site pain, swelling, arm motion limitation, headache, and malaise were increased in the vaccine groups. There was no evidence for effect of the CS-NANP vaccines (307 participants, 3 trials), CS102 peptide vaccine (14 participants, 1 trial), or the ME-TRAP vaccine (372 participants, 1 trial). AUTHORS' CONCLUSIONS RTS,S vaccine was effective in preventing a significant number of clinical malaria episodes, including good protection against severe malaria in children for 18 months. No severe adverse events were attributable to the vaccine. Progression of this vaccine towards licensing is justified while efforts to increase its efficacy continue. The other vaccines do not look promising and further research is a priority.
Collapse
Affiliation(s)
- P Graves
- EpiVec Consulting, 606 Kimberly Lane NE, Atlanta, GA 30306, USA.
| | | |
Collapse
|
40
|
Im EJ, Nkolola JP, di Gleria K, McMichael AJ, Hanke T. Induction of long-lasting multi-specific CD8+T cells by a four-component DNA-MVA/HIVA-RENTA candidate HIV-1 vaccine in rhesus macaques. Eur J Immunol 2006; 36:2574-84. [PMID: 17013988 DOI: 10.1002/eji.200636482] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
As a part of a long-term effort to develop vaccine against HIV-1 clade A inducing protective T cell responses in humans, we run mutually complementing studies in humans and non-human primates (NHP) with the aim to maximize vaccine immunogenicity. The candidate vaccine under development has four components, pTHr.HIVA and pTH.RENTA DNA, and modified vaccinia virus Ankara (MVA).HIVA and MVA.RENTA, delivered in a heterologous DNA prime-MVA boost regimen. While the HIVA (Gag/epitopes) components have been tested in NHP and over 300 human subjects, we plan to test in humans the RENTA (reverse transcriptase, gp41, Nef, Tat) vaccines designed to broaden HIVA-induced responses in year 2007. Here, we investigated the four-component vaccine long-term immunogenicity in Mamu-A*01-positive rhesus macaques and demonstrated that the vaccine-induced T cells were multi-specific, multi-functional, readily proliferated to recall peptides and were circulating in the peripheral blood of vaccine recipients over 1 year after vaccine administration. The consensus clade A-elicited T cells recognized 50% of tested epitope variants from other HIV-1 clades. Thus, the DNA-MVA/HIVA-RENTA vaccine induced memory T cells of desirable characteristics and similarities to those induced in humans by HIVA vaccines alone; however, single-clade vaccines may not elicit sufficiently cross-reactive responses.
Collapse
Affiliation(s)
- Eung-Jun Im
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | | | | | | | | |
Collapse
|
41
|
Bejon P, Keating S, Mwacharo J, Kai OK, Dunachie S, Walther M, Berthoud T, Lang T, Epstein J, Carucci D, Moris P, Cohen J, Gilbert SC, Peshu N, Marsh K, Hill AVS. Early gamma interferon and interleukin-2 responses to vaccination predict the late resting memory in malaria-naïve and malaria-exposed individuals. Infect Immun 2006; 74:6331-8. [PMID: 16966412 PMCID: PMC1695504 DOI: 10.1128/iai.00774-06] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Two different cell populations respond to potent T-cell-inducing vaccinations. The induction and loss of effector cells can be seen using an ex vivo enzyme-linked immunospot (ELISPOT) assay, but the more durable resting memory response is demonstrable by a cultured ELISPOT assay. The relationship of the early effector response to durable resting memory is incompletely understood. Effector phenotype is usually identified by gamma interferon (IFN-gamma) production, but interleukin-2 (IL-2) has been specifically linked to the differentiation of memory cells. Here, IFN-gamma- and IL-2-secreting effector cells were identified by an ex vivo ELISPOT assay 1 week after vaccination and compared with the resting memory responses detected by a cultured ELISPOT assay 3 months later. The different kinetics and induction of IL-2 by different vaccines and natural exposure are described. Furthermore, both early IFN-gamma and IL-2 production independently predicted subsequent memory responses at 3 months in malaria-naïve volunteers, but only IFN-gamma predicted memory in malaria-exposed volunteers. However, dual ELISPOT assays were also performed on malaria-exposed volunteers to identify cells producing both cytokines simultaneously. This demonstrated that double-cytokine-producing cells were highly predictive of memory. This assay may be useful in predicting vaccinations most likely to generate stable, long-term memory responses.
Collapse
Affiliation(s)
- Philip Bejon
- Kenya Medical Research Institute, Centre for Geographical Medical Research (Coast), P.O. Box 230, Kilifi, Kenya.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Mulligan MJ, Russell ND, Celum C, Kahn J, Noonan E, Montefiori DC, Ferrari G, Weinhold KJ, Smith JM, Amara RR, Robinson HL. Excellent safety and tolerability of the human immunodeficiency virus type 1 pGA2/JS2 plasmid DNA priming vector vaccine in HIV type 1 uninfected adults. AIDS Res Hum Retroviruses 2006; 22:678-83. [PMID: 16831092 DOI: 10.1089/aid.2006.22.678] [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] [Indexed: 11/12/2022] Open
Abstract
A vaccine consisting of DNA priming followed by recombinant modified vaccinia Ankara (rMVA) boosting has achieved long-term control of a pathogenic challenge with a chimera of simian and human immunodeficiency viruses (SHIV-89.6P) in rhesus macaques. Based on these results, clade B HIV-1 DNA and rMVA immunogens have been developed for trials in humans. We conducted a first-time in humans phase I safety trial using the pGA2/JS2 (JS2) HIV-1 DNA priming vector expressing Gag, Pol, Env, Tat, Rev, and Vpu. Thirty HIV-uninfected adults were vaccinated with 0.3 or 3 mg of JS2 DNA, or a saline placebo, by intramuscular injection at months 0 and 2. Both doses of DNA were safe and well-tolerated with no differences between the control, 0.3 mg, or 3 mg groups (n = 6, 12, and 12, respectively) through 12 months of postvaccination follow- up. A chromium-release assay using fresh peripheral blood mononuclear cells (PBMCs) and a validated IFN-gamma ELISpot assay with frozen PBMCs failed to detect CD4(+) or CD8(+) HIV-1-specific T cell responses. HIV-specific neutralizing antibodies were also not detected. The vaccine is being further developed as a priming vector for a combined DNA plus rMVA prime/boost HIV vaccination regimen.
Collapse
|
43
|
Walther M. Advances in vaccine development against the pre-erythrocytic stage of Plasmodium falciparum malaria. Expert Rev Vaccines 2006; 5:81-93. [PMID: 16451110 DOI: 10.1586/14760584.5.1.81] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
With approximately 2.4 billion people at risk, Plasmodium falciparum malaria infection caused by an infectious bite of an Anopheles mosquito continues to be a major cause of mortality and morbidity, mainly in the tropics and subtropics. Measures to control the mosquito vector on a broad scale are expensive and need to be maintained continuously. The rapid emergence of parasite strains that are resistant to affordable drugs highlights the urgent need for a cheap and effective vaccine. Candidate vaccines that have been developed to date target different stages of the parasite life cycle. This review describes the recent advances in the development of a vaccine that aims to terminate the infection at its first stage in the liver. The candidate vaccines that are currently under clinical evaluation are introduced and the results from recent trials discussed. The review aims to explain the immunologic challenges a successful vaccine has to meet, as well as the different strategies that are currently employed in an attempt to induce a protective immune response. Furthermore, an outline of available options to be tested in the near future will be presented.
Collapse
Affiliation(s)
- Michael Walther
- MRC Laboratories, Fajara PO Box 273, Banjul, West Africa, The Gambia.
| |
Collapse
|
44
|
Imoukhuede EB, Berthoud T, Milligan P, Bojang K, Ismaili J, Keating S, Nwakanma D, Keita S, Njie F, Sowe M, Todryk S, Laidlaw SM, Skinner MA, Lang T, Gilbert S, Greenwood BM, Hill AVS. Safety and immunogenicity of the malaria candidate vaccines FP9 CS and MVA CS in adult Gambian men. Vaccine 2006; 24:6526-33. [PMID: 16842888 DOI: 10.1016/j.vaccine.2006.06.022] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2006] [Revised: 06/06/2006] [Accepted: 06/14/2006] [Indexed: 11/26/2022]
Abstract
We assessed the safety and immunogenicity of prime-boost vectors encoding the Plasmodium falciparum circumsporozoite (CS) protein expressed either in the attenuated fowl-pox virus (FP9) or modified vaccinia virus Ankara (MVA). Thirty-two adult Gambians in groups of four to eight received one, two or three doses of FP9 CS and/or MVA CS. No serious adverse event was observed following vaccination. The most immunogenic regimen was two doses of FP9 followed by a single dose of MVA 4 weeks later (an average of 1000 IFN-gamma spot forming units/million PBMCs). This level of effector T-cell responses appears higher than that seen in previously reported studies of CS-based candidate malaria vaccines.
Collapse
|
45
|
Sirima SB, Nébié I, Ouédraogo A, Tiono AB, Konaté AT, Gansané A, Dermé AI, Diarra A, Ouédraogo A, Soulama I, Cuzzin-Ouattara N, Cousens S, Leroy O. Safety and immunogenicity of the Plasmodium falciparum merozoite surface protein-3 long synthetic peptide (MSP3-LSP) malaria vaccine in healthy, semi-immune adult males in Burkina Faso, West Africa. Vaccine 2006; 25:2723-32. [PMID: 17280744 DOI: 10.1016/j.vaccine.2006.05.090] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2006] [Revised: 05/18/2006] [Accepted: 05/23/2006] [Indexed: 11/15/2022]
Abstract
UNLABELLED The merozoite surface protein-3 long synthetic peptide (MSP3-LSP) comprises the amino acid sequence 186-276 of the Plasmodium falciparum protein MSP3. It is currently in development as an erythrocytic stage (blood stage) malaria vaccine candidate. We report here the first data on the safety, reactogenicity and immunogenicity of three doses of MSP3-LSP, adjuvanted with aluminium hydroxide, in healthy male adults living in a malaria endemic area. METHODS A phase 1b single-blind controlled trial was performed in the village of Balonghin in Burkina Faso. Thirty male volunteers aged 18-40 years were randomised to receive either three doses of 30 microg MSP3-LSP or 0.5 ml of tetanus toxoid vaccine. The second and third vaccine doses were given 28 and 112 days after the first dose. We followed participants for 1 year. RESULTS There were no serious adverse events in either vaccine group. In both groups participants reported local reactions at the site of injection when compared to an earlier trial in European volunteers. Only one systemic adverse event (tachycardia) was identified which occurred immediately after the first vaccination in one individual receiving MSP3-LSP. No clinically significant biological abnormalities following vaccination were observed. Humoral immune responses (IgG, IgG subclasses, IgM) to MSP3-LSP peptide were similar in the two groups following vaccination. Some cell-mediated immune responses appeared to differ between the two vaccine groups. After the second dose of MSP3-LSP, there appeared to be a marked increase in the lymphocyte proliferation index and IFN-gamma in response to stimulation with MSP3-LSP. CONCLUSION These data suggest that three doses of 30 microg MSP3-LSP when administered subcutaneously on days 0, 28 and 112 are well-tolerated by adult males previously exposed to natural P. falciparum infection. They also suggest that MSP3-LSP is able to stimulate an enhanced cell-mediated immune response in individuals with some degree of preexisting immunity.
Collapse
Affiliation(s)
- Sodiomon B Sirima
- Centre National de Recherche et de Formation sur le Paludisme (CNRFP), 01 BP 2208 Ouagadougou 01, Burkina Faso.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Goonetilleke N, Moore S, Dally L, Winstone N, Cebere I, Mahmoud A, Pinheiro S, Gillespie G, Brown D, Loach V, Roberts J, Guimaraes-Walker A, Hayes P, Loughran K, Smith C, De Bont J, Verlinde C, Vooijs D, Schmidt C, Boaz M, Gilmour J, Fast P, Dorrell L, Hanke T, McMichael AJ. Induction of multifunctional human immunodeficiency virus type 1 (HIV-1)-specific T cells capable of proliferation in healthy subjects by using a prime-boost regimen of DNA- and modified vaccinia virus Ankara-vectored vaccines expressing HIV-1 Gag coupled to CD8+ T-cell epitopes. J Virol 2006; 80:4717-28. [PMID: 16641265 PMCID: PMC1472051 DOI: 10.1128/jvi.80.10.4717-4728.2006] [Citation(s) in RCA: 195] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
A double-blind randomized phase I trial was conducted in human immunodeficiency virus type 1 (HIV-1)-negative subjects receiving vaccines vectored by plasmid DNA and modified vaccinia virus Ankara (MVA) expressing HIV-1 p24/p17 gag linked to a string of CD8(+) T-cell epitopes. The trial had two groups. One group received either two doses of MVA.HIVA (2x MVA.HIVA) (n=8) or two doses of placebo (2x placebo) (n=4). The second group received 2x pTHr.HIVA followed by one dose of MVA.HIVA (n=8) or 3x placebo (n=4). In the pTHr.HIVA-MVA.HIVA group, HIV-1-specific T-cell responses peaked 1 week after MVA.HIVA vaccination in both ex vivo gamma interferon (IFN-gamma) ELISPOT (group mean, 210 spot-forming cells/10(6) cells) and proliferation (group mean stimulation index, 37), with assays detecting positive responses in four out of eight and five out of eight subjects, respectively. No HIV-1-specific T-cell responses were detected in either assay in the 2x MVA.HIVA group or subjects receiving placebo. Using a highly sensitive and reproducible cultured IFN-gamma ELISPOT assay, positive responses mainly mediated by CD4(+) T cells were detected in eight out of eight vaccinees in the pTHr.HIVA-MVA.HIVA group and four out of eight vaccinees in the 2x MVA.HIVA group. Importantly, no false-positive responses were detected in the eight subjects receiving placebo. Of the 12 responders, 11 developed responses to previously identified immunodominant CD4(+) T-cell epitopes, with 6 volunteers having responses to more than one epitope. Five out of 12 responders also developed CD8(+) T-cell responses to the epitope string. Induced T cells produced a variety of anti-viral cytokines, including tumor necrosis factor alpha and macrophage inflammatory protein 1 beta. These data demonstrate that prime-boost vaccination with recombinant DNA and MVA vectors can induce multifunctional HIV-1-specific T cells in the majority of vaccinees.
Collapse
MESH Headings
- AIDS Vaccines/genetics
- AIDS Vaccines/immunology
- Amino Acid Sequence
- CD8-Positive T-Lymphocytes/cytology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Cell Proliferation
- Cells, Cultured
- Double-Blind Method
- Epitopes, T-Lymphocyte/immunology
- Epitopes, T-Lymphocyte/metabolism
- Gene Products, gag/immunology
- Gene Products, gag/metabolism
- Genetic Vectors
- HIV Infections/prevention & control
- HIV-1/genetics
- HIV-1/immunology
- Humans
- Immunization, Secondary
- Lymphocyte Activation/immunology
- Molecular Sequence Data
- Vaccines, DNA/genetics
- Vaccines, DNA/immunology
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
- Vaccinia virus/genetics
- Vaccinia virus/immunology
Collapse
Affiliation(s)
- Nilu Goonetilleke
- Centre for Clinical Vaccinology and Tropical Medicine and MRC Human Immunology Unit, University of Oxford, Oxford OX3 7LJ, United Kingdom.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Miao J, Li X, Liu Z, Xue C, Bujard H, Cui L. Immune responses in mice induced by prime-boost schemes of the Plasmodium falciparum apical membrane antigen 1 (PfAMA1)-based DNA, protein and recombinant modified vaccinia Ankara vaccines. Vaccine 2006; 24:6187-98. [PMID: 16806600 DOI: 10.1016/j.vaccine.2006.05.099] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2006] [Revised: 05/25/2006] [Accepted: 05/31/2006] [Indexed: 10/24/2022]
Abstract
The apical membrane antigen 1 (AMA1) of malaria parasites is a leading vaccine candidate. Its expression in merozoites and sporozoites and its importance for erythrocyte and hepatocyte invasion underline the significance of both humoral and cellular immunities against this antigen in malaria protection. We have generated a DNA construct and a recombinant poxvirus (rMVA) for expressing the Plasmodium falciparum AMA1 ectodomain, produced recombinant AMA1 protein (rAMA1) and evaluated their antigenicity in mice using single and combinatory vaccine schemes. Our results showed that although vaccinations of mice by either DNA or rMVA alone did not yield high antibody responses, they had primed significant numbers of rAMA1-responsive splenocytes. Under heterologous prime-boost schemes, priming with DNA followed by boosting with rMVA or rAMA1 protein resulted in a significant increase in antibody titers. In addition, the antibody titers to AMA1 appeared to be correlated with the levels of inhibition of merozoite invasion of erythrocytes in vitro. Furthermore, different prime-boost schemes resulted in different AMA1-specific antibody isotype (IgG1/IgG2a) ratios, providing us with an indication about Th1 or Th2 responses the vaccination regimens have induced. This study has yielded useful information for further in vivo evaluation of the suitability and effectiveness of the heterologous prime-boost strategy in AMA1 vaccination.
Collapse
MESH Headings
- Animals
- Antigens, Protozoan/genetics
- Antigens, Protozoan/immunology
- Chick Embryo
- Cricetinae
- HeLa Cells
- Humans
- Immunization, Secondary
- Malaria Vaccines/genetics
- Malaria Vaccines/immunology
- Malaria Vaccines/therapeutic use
- Malaria, Falciparum/immunology
- Malaria, Falciparum/parasitology
- Membrane Proteins/genetics
- Membrane Proteins/immunology
- Mice
- Mice, Inbred BALB C
- Plasmodium falciparum/genetics
- Plasmodium falciparum/immunology
- Protozoan Proteins/genetics
- Protozoan Proteins/immunology
- Rabbits
- Th1 Cells/immunology
- Vaccines, DNA/genetics
- Vaccines, DNA/immunology
- Vaccines, Subunit/genetics
- Vaccines, Subunit/immunology
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
- Vaccinia virus/genetics
- Vaccinia virus/immunology
Collapse
Affiliation(s)
- Jun Miao
- Department of Entomology, The Pennsylvania State University, 501 ASI Building, University Park, PA 16802, USA
| | | | | | | | | | | |
Collapse
|
48
|
Walther M, Thompson FM, Dunachie S, Keating S, Todryk S, Berthoud T, Andrews L, Andersen RF, Moore A, Gilbert SC, Poulton I, Dubovsky F, Tierney E, Correa S, Huntcooke A, Butcher G, Williams J, Sinden RE, Hill AVS. Safety, immunogenicity, and efficacy of prime-boost immunization with recombinant poxvirus FP9 and modified vaccinia virus Ankara encoding the full-length Plasmodium falciparum circumsporozoite protein. Infect Immun 2006; 74:2706-16. [PMID: 16622207 PMCID: PMC1459746 DOI: 10.1128/iai.74.5.2706-2716.2006] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Heterologous prime-boost immunization with DNA and various recombinant poxviruses encoding malaria antigens is capable of inducing strong cell-mediated immune responses and partial protection in human sporozoite challenges. Here we report a series of trials assessing recombinant fowlpox virus and modified vaccinia virus Ankara encoding the Plasmodium falciparum circumsporozoite protein in various prime-boost combinations, doses, and application routes. For the first time, these vaccines were administered intramuscularly and at doses of up to 5 x 10(8) PFU. Vaccines containing this antigen proved safe and induced modest immune responses but showed no evidence of efficacy in a sporozoite challenge.
Collapse
Affiliation(s)
- Michael Walther
- Centre for Clinical Vaccinology & Tropical Medicine, Nuffield Department of Clinical Medicine, Oxford University, United Kingdom.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
Persistent hepatitis B virus (HBV) infection represents a major public health concern because of its association with chronic liver disease and the propensity of the disease to progress to cirrhosis and hepatocellular carcinoma. Despite the availability of a prophylactic vaccine effective in a majority of the population, alternative vaccination strategies are being sought to induce protective responses in healthy non-responders and to boost and broaden T cell responses in chronically infected patients, which may lead to a better control of the virus and/or its eventual complete clearance. In this issue of the European Journal of Immunology immunization of BALB/c mice intramuscularly with a DNA vaccine encoding the hepatitis virus B surface antigen (HBsAg) was shown to result in prolonged secretion of HBsAg into the serum and the elicitation of HBsAg-specific antibodies. In fact, the vaccine was so efficient that the antibodies and HBsAg formed circulating immune complexes and induced kidney and liver lesions similar to those observed in chronically infected patients. This commentary discusses these results in terms of the safety of plasmid DNA-vectored genetic vaccines in general, the use of DNA vaccines expressing HBsAg for the treatment of chronic hepatitis and the consequences of prolonged immunogen expression for the development of protective immune responses.
Collapse
Affiliation(s)
- Tomás Hanke
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
50
|
Gilbert SC, Moorthy VS, Andrews L, Pathan AA, McConkey SJ, Vuola JM, Keating SM, Berthoud T, Webster D, McShane H, Hill AVS. Synergistic DNA–MVA prime-boost vaccination regimes for malaria and tuberculosis. Vaccine 2006; 24:4554-61. [PMID: 16150517 DOI: 10.1016/j.vaccine.2005.08.048] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
T-cell-mediated responses against the liver-stage of Plasmodium falciparum are critical for protection in the human irradiated sporozoite model and several animal models. Heterologous prime-boost approaches, employing plasmid DNA and viral vector delivery of malarial DNA sequences, have proved particularly promising for maximising T-cell-mediated protection in animal models. The T-cell responses induced by this prime-boost regime, in animals and humans, are substantially greater than the sum of the responses induced by DNA or MVA vaccines used alone, leading to the term introduced here of "synergistic" prime-boost immunisation. The insert in our first generation clinical constructs is known as multiple epitope-thrombospondin-related adhesion protein (ME-TRAP). We have performed an extensive series of phase I/II trials evaluating various prime-boost combination regimens for delivery of ME-TRAP in over 500 malaria-naïve and malaria-exposed individuals. The three delivery vectors are DNA, modified vaccinia virus Ankara (MVA) and, more recently, fowlpox strain 9 (FP9). Administration was intra-epidermal and intramuscular for DNA and intradermal for MVA and FP9. Doses of DNA ranged from 4 microg to 2mg. Doses of MVA were up to 1.5 x 10(8) plaque forming units (pfu) and of FP9, up to 1.0 x 10(8)pfu. Further trials employing bacille Calmette-Guérin (BCG) as the priming agent and MVA expressing antigen 85A of Mycobacterium tuberculosis as the boosting agent has extended the scope of synergistic prime-boost vaccination. In this review we summarise the safety, immunogenicity and efficacy results from these malaria and tuberculosis vaccine clinical trials.
Collapse
Affiliation(s)
- Sarah C Gilbert
- Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|