1
|
Silva-Pilipich N, Beloki U, Salaberry L, Smerdou C. Self-Amplifying RNA: A Second Revolution of mRNA Vaccines against COVID-19. Vaccines (Basel) 2024; 12:318. [PMID: 38543952 PMCID: PMC10974399 DOI: 10.3390/vaccines12030318] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/11/2024] [Accepted: 03/12/2024] [Indexed: 11/12/2024] Open
Abstract
SARS-CoV-2 virus, the causative agent of COVID-19, has produced the largest pandemic in the 21st century, becoming a very serious health problem worldwide. To prevent COVID-19 disease and infection, a large number of vaccines have been developed and approved in record time, including new vaccines based on mRNA encapsulated in lipid nanoparticles. While mRNA-based vaccines have proven to be safe and effective, they are more expensive to produce compared to conventional vaccines. A special type of mRNA vaccine is based on self-amplifying RNA (saRNA) derived from the genome of RNA viruses, mainly alphaviruses. These saRNAs encode a viral replicase in addition to the antigen, usually the SARS-CoV-2 spike protein. The replicase can amplify the saRNA in transfected cells, potentially reducing the amount of RNA needed for vaccination and promoting interferon I responses that can enhance adaptive immunity. Preclinical studies with saRNA-based COVID-19 vaccines in diverse animal models have demonstrated the induction of robust protective immune responses, similar to conventional mRNA but at lower doses. Initial clinical trials have confirmed the safety and immunogenicity of saRNA-based vaccines in individuals that had previously received authorized COVID-19 vaccines. These findings have led to the recent approval of two of these vaccines by the national drug agencies of India and Japan, underscoring the promising potential of this technology.
Collapse
Affiliation(s)
- Noelia Silva-Pilipich
- Division of DNA and RNA Medicine, Cima Universidad de Navarra, 31008 Pamplona, Spain;
- Instituto de Investigación Sanitaria de Navarra (IdISNA) and CCUN, 31008 Pamplona, Spain
| | - Uxue Beloki
- Division of DNA and RNA Medicine, Cima Universidad de Navarra, 31008 Pamplona, Spain;
- Instituto de Investigación Sanitaria de Navarra (IdISNA) and CCUN, 31008 Pamplona, Spain
| | - Laura Salaberry
- Facultad de Ingeniería, Universidad ORT Uruguay, Montevideo 11100, Uruguay;
- Nanogrow Biotech, Montevideo 11500, Uruguay
| | - Cristian Smerdou
- Division of DNA and RNA Medicine, Cima Universidad de Navarra, 31008 Pamplona, Spain;
- Instituto de Investigación Sanitaria de Navarra (IdISNA) and CCUN, 31008 Pamplona, Spain
| |
Collapse
|
2
|
Potent Anti-hepatitis C Virus (HCV) T Cell Immune Responses Induced in Mice Vaccinated with DNA-Launched RNA Replicons and Modified Vaccinia Virus Ankara-HCV. J Virol 2019; 93:JVI.00055-19. [PMID: 30674625 DOI: 10.1128/jvi.00055-19] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 01/11/2019] [Indexed: 02/06/2023] Open
Abstract
Hepatitis C is a liver disease caused by the hepatitis C virus (HCV) affecting 71 million people worldwide with no licensed vaccines that prevent infection. Here, we have generated four novel alphavirus-based DNA-launched self-amplifying RNA replicon (DREP) vaccines expressing either structural core-E1-E2 or nonstructural p7-NS2-NS3 HCV proteins of genotype 1a placed under the control of an alphavirus promoter, with or without an alphaviral translational enhancer (grouped as DREP-HCV or DREP-e-HCV, respectively). DREP vectors are known to induce cross-priming and further stimulation of immune responses through apoptosis, and here we demonstrate that they efficiently trigger apoptosis-related proteins in transfected cells. Immunization of mice with the DREP vaccines as the priming immunization followed by a heterologous boost with a recombinant modified vaccinia virus Ankara (MVA) vector expressing the nearly full-length genome of HCV (MVA-HCV) induced potent and long-lasting HCV-specific CD4+ and CD8+ T cell immune responses that were significantly stronger than those of a homologous MVA-HCV prime/boost immunization, with the DREP-e-HCV/MVA-HCV combination the most immunogenic regimen. HCV-specific CD4+ and CD8+ T cell responses were highly polyfunctional, had an effector memory phenotype, and were mainly directed against E1-E2 and NS2-NS3, respectively. Additionally, DREP/MVA-HCV immunization regimens induced higher antibody levels against HCV E2 protein than homologous MVA-HCV immunization. Collectively, these results provided an immunization protocol against HCV by inducing high levels of HCV-specific T cell responses as well as humoral responses. These findings reinforce the combined use of DREP-based vectors and MVA-HCV as promising prophylactic and therapeutic vaccines against HCV.IMPORTANCE HCV represents a global health problem as more than 71 million people are chronically infected worldwide. Direct-acting antiviral agents can cure HCV infection in most patients, but due to the high cost of these agents and the emergence of resistant mutants, they do not represent a feasible and affordable strategy to eradicate the virus. Therefore, a vaccine is an urgent goal that requires efforts to understand the correlates of protection for HCV clearance. Here, we describe for the first time the generation of novel vaccines against HCV based on alphavirus DNA replicons expressing HCV antigens. We demonstrate that potent T cell immune responses, as well as humoral immune responses, against HCV can be achieved in mice by using a combined heterologous prime/boost immunization protocol consisting of the administration of alphavirus replicon DNA vectors as the priming immunization followed by a boost with a recombinant modified vaccinia virus Ankara vector expressing HCV antigens.
Collapse
|
3
|
van de Wall S, Ljungberg K, Ip PP, Boerma A, Knudsen ML, Nijman HW, Liljeström P, Daemen T. Potent therapeutic efficacy of an alphavirus replicon DNA vaccine expressing human papilloma virus E6 and E7 antigens. Oncoimmunology 2018; 7:e1487913. [PMID: 30288352 PMCID: PMC6169581 DOI: 10.1080/2162402x.2018.1487913] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 06/06/2018] [Accepted: 06/06/2018] [Indexed: 12/22/2022] Open
Abstract
Cervical cancer develops as a result of infection with high-risk human papillomavirus (HPV) through persistent expression of early proteins E6 and E7. Our group pioneered a recombinant viral vector system based on Semliki Forest virus (SFV) for vaccination against cervical cancer. The most striking benefit of this alphavirus vector-based vaccine platform is its high potency. DNA vaccines on the other hand, have a major advantage with respect to ease of production. In this study, the benefits associated with both SFV-based vaccines and DNA vaccines were combined with the development of a DNA-launched RNA replicon (DREP) vaccine targeting cervical cancer. Using intradermal delivery followed by electroporation, we demonstrated that DREP encoding for E6,7 (DREP-E6,7) induced effective, therapeutic antitumor immunity. While immunizations with a conventional DNA vaccine did not prevent tumor outgrowth, immunization with a 200-fold lower equimolar dose of DREP (0.05 µg of DREP) resulted in approximately 85% of tumor-free mice. To overcome the safety concern of potential malignant transformation at the vaccination site, we evaluated the anti-tumor effect of a DREP vaccine encoding a shuffled version of E7 (DREP-E7sh). DREP-E7sh delayed tumor growth yet not to the same extent as DREP-E6,7. In addition, inclusion of a helper cassette and an ER targeting signal (sigHelp) did not significantly further enhance the suppression of tumor outgrowth in the long term, albeit exhibiting better tumor control early after immunization. Collectively, this study points towards the clinical evaluation of DREP encoding HPV antigens as a potent immunotherapy for patients with HPV16 (pre)-malignancies.
Collapse
Affiliation(s)
- Stephanie van de Wall
- Department of Medical Microbiology, Tumor Virology and Cancer Immunotherapy, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Karl Ljungberg
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Peng Peng Ip
- Department of Medical Microbiology, Tumor Virology and Cancer Immunotherapy, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Annemarie Boerma
- Department of Medical Microbiology, Tumor Virology and Cancer Immunotherapy, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Maria L Knudsen
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Hans W Nijman
- Department of Obstetrics & Gynecology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Peter Liljeström
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Toos Daemen
- Department of Medical Microbiology, Tumor Virology and Cancer Immunotherapy, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
4
|
Ren S, Wei Q, Cai L, Yang X, Xing C, Tan F, Leavenworth JW, Liang S, Liu W. Alphavirus Replicon DNA Vectors Expressing Ebola GP and VP40 Antigens Induce Humoral and Cellular Immune Responses in Mice. Front Microbiol 2018; 8:2662. [PMID: 29375526 PMCID: PMC5767729 DOI: 10.3389/fmicb.2017.02662] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 12/20/2017] [Indexed: 11/13/2022] Open
Abstract
Ebola virus (EBOV) causes severe hemorrhagic fevers in humans, and no approved therapeutics or vaccine is currently available. Glycoprotein (GP) is the major protective antigen of EBOV, and can generate virus-like particles (VLPs) by co-expression with matrix protein (VP40). In this study, we constructed a recombinant Alphavirus Semliki Forest virus (SFV) replicon vector DREP to express EBOV GP and matrix viral protein (VP40). EBOV VLPs were successfully generated and achieved budding from 293 cells after co-transfection with DREP-based GP and VP40 vectors (DREP-GP+DREP-VP40). Vaccination of BALB/c mice with DREP-GP, DREP-VP40, or DREP-GP+DREP-VP40 vectors, followed by immediate electroporation resulted in a mixed IgG subclass production, which recognized EBOV GP and/or VP40 proteins. This vaccination regimen also led to the generation of both Th1 and Th2 cellular immune responses in mice. Notably, vaccination with DREP-GP and DREP-VP40, which produces both GP and VP40 antigens, induced a significantly higher level of anti-GP IgG2a antibody and increased IFN-γ secreting CD8+ T-cell responses relative to vaccination with DREP-GP or DREP-VP40 vector alone. Our study indicates that co-expression of GP and VP40 antigens based on the SFV replicon vector generates EBOV VLPs in vitro, and vaccination with recombinant DREP vectors containing GP and VP40 antigens induces Ebola antigen-specific humoral and cellular immune responses in mice. This novel approach provides a simple and efficient vaccine platform for Ebola disease prevention.
Collapse
Affiliation(s)
- Shoufeng Ren
- Department of Human Parasitology, Wenzhou Medical University, Wenzhou, China
| | - Qimei Wei
- Department of Human Parasitology, Wenzhou Medical University, Wenzhou, China.,Institute of Pathogen and Immunology, Wenzhou Medical University, Wenzhou, China
| | - Liya Cai
- Department of Human Parasitology, Wenzhou Medical University, Wenzhou, China.,Institute of Pathogen and Immunology, Wenzhou Medical University, Wenzhou, China
| | - Xuejing Yang
- Department of Laboratory Medicine, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Cuicui Xing
- Department of Human Parasitology, Wenzhou Medical University, Wenzhou, China
| | - Feng Tan
- Department of Human Parasitology, Wenzhou Medical University, Wenzhou, China.,Institute of Pathogen and Immunology, Wenzhou Medical University, Wenzhou, China
| | - Jianmei W Leavenworth
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, United States.,Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Shaohui Liang
- Department of Human Parasitology, Wenzhou Medical University, Wenzhou, China.,Institute of Pathogen and Immunology, Wenzhou Medical University, Wenzhou, China
| | - Wenquan Liu
- Department of Human Parasitology, Wenzhou Medical University, Wenzhou, China.,Institute of Pathogen and Immunology, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
5
|
Protective immune response in mice induced by a suicidal DNA vaccine encoding NTPase-II gene of Toxoplasma gondii. Acta Trop 2017; 166:336-342. [PMID: 27940233 DOI: 10.1016/j.actatropica.2016.12.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 10/24/2016] [Accepted: 12/05/2016] [Indexed: 11/24/2022]
Abstract
DNA-based alphaviral RNA replicon vectors, also called suicidal DNA vectors, have been employed to alleviate biosafety concerns attribution to its ability to induce apoptotic cell death of the transfected cells. Toxoplasma gondii nucleoside triphosphate hydrolase-II (TgNTPase-II), which facilitates the parasite to salvage purines from the host cell for survival and replication, have been demonstrated to be a potential vaccine candidate for toxoplasmosis. Herein, we evaluated the immunogenic potential of a suicidal DNA vaccine encoding TgNTPase-II gene, pDREP-TgNTPase-II, delivered intramuscularly in combination with electroporation. Immunization of mice with pDREP-TgNTPase-II elicited specific humoral responses, with high IgG antibody titers and a mixed IgG1/IgG2a response. The cellular immune response was associated with high level production of IFN-γ, IL-2, IL-10 cytokines and low level IL-4 production as well as the increase of the percentage of CD8+ T cells, indicating that a Th1 predominant response was elicited. Furthermore, mice vaccinated with this suicidal DNA vaccine displayed partial protection against acute infection with the virulent RH strain as well as chronic infection with PRU cyst, which shows 77.7% and 71.4% reduction in brain cyst burden in comparison to PBS and pDREP-eGFP control group, respectively. Based on the cellular and antibody responses, the suicidal DNA vaccine elicited a Th1-predominant immune response against T. gondii challenge.
Collapse
|
6
|
Knudsen ML, Ljungberg K, Tatoud R, Weber J, Esteban M, Liljeström P. Alphavirus replicon DNA expressing HIV antigens is an excellent prime for boosting with recombinant modified vaccinia Ankara (MVA) or with HIV gp140 protein antigen. PLoS One 2015; 10:e0117042. [PMID: 25643354 PMCID: PMC4314072 DOI: 10.1371/journal.pone.0117042] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 12/18/2014] [Indexed: 12/31/2022] Open
Abstract
Vaccination with DNA is an attractive strategy for induction of pathogen-specific T cells and antibodies. Studies in humans have shown that DNA vaccines are safe, but their immunogenicity needs further improvement. As a step towards this goal, we have previously demonstrated that immunogenicity is increased with the use of an alphavirus DNA-launched replicon (DREP) vector compared to conventional DNA vaccines. In this study, we investigated the effect of varying the dose and number of administrations of DREP when given as a prime prior to a heterologous boost with poxvirus vector (MVA) and/or HIV gp140 protein formulated in glucopyranosyl lipid A (GLA-AF) adjuvant. The DREP and MVA vaccine constructs encoded Env and a Gag-Pol-Nef fusion protein from HIV clade C. One to three administrations of 0.2 μg DREP induced lower HIV-specific T cell and IgG responses than the equivalent number of immunizations with 10 μg DREP. However, the two doses were equally efficient as a priming component in a heterologous prime-boost regimen. The magnitude of immune responses depended on the number of priming immunizations rather than the dose. A single low dose of DREP prior to a heterologous boost resulted in greatly increased immune responses compared to MVA or protein antigen alone, demonstrating that a mere 0.2 μg DREP was sufficient for priming immune responses. Following a DREP prime, T cell responses were expanded greatly by an MVA boost, and IgG responses were also expanded when boosted with protein antigen. When MVA and protein were administered simultaneously following multiple DREP primes, responses were slightly compromised compared to administering them sequentially. In conclusion, we have demonstrated efficient priming of HIV-specific T cell and IgG responses with a low dose of DREP, and shown that the priming effect depends on number of primes administered rather than dose.
Collapse
MESH Headings
- Alphavirus/genetics
- Animals
- Antibodies, Viral/immunology
- Chemistry, Pharmaceutical
- DNA, Recombinant/genetics
- DNA, Viral/genetics
- Female
- Gene Expression
- Genetic Vectors/genetics
- HIV Antigens/genetics
- HIV Antigens/immunology
- HIV-1/immunology
- Immunization, Secondary
- Immunoglobulin G/immunology
- Lipid A/chemistry
- Mice
- Mice, Inbred BALB C
- Replicon/genetics
- T-Lymphocytes/immunology
- Vaccines, DNA/genetics
- Vaccines, DNA/immunology
- Vaccinia virus/genetics
- env Gene Products, Human Immunodeficiency Virus/chemistry
- env Gene Products, Human Immunodeficiency Virus/immunology
Collapse
Affiliation(s)
- Maria L. Knudsen
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
- * E-mail: (MLK); (PL)
| | - Karl Ljungberg
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Roger Tatoud
- Imperial College London, Department of Infectious Diseases, Division of Medicine, Norfolk Place, London, United Kingdom
| | - Jonathan Weber
- Imperial College London, Department of Infectious Diseases, Division of Medicine, Norfolk Place, London, United Kingdom
| | - Mariano Esteban
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Peter Liljeström
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
- * E-mail: (MLK); (PL)
| |
Collapse
|
7
|
Wang HC, An HJ, Yu YZ, Xu Q. Potentiation of anthrax vaccines using protective antigen-expressing viral replicon vectors. Immunol Lett 2015; 163:206-13. [DOI: 10.1016/j.imlet.2014.07.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 07/18/2014] [Accepted: 07/27/2014] [Indexed: 11/17/2022]
|
8
|
Abstract
UNLABELLED Chikungunya virus (CHIKV) is a reemerging mosquito-borne alphavirus that causes debilitating arthralgia in humans. Here we describe the development and testing of novel DNA replicon and protein CHIKV vaccine candidates and evaluate their abilities to induce antigen-specific immune responses against CHIKV. We also describe homologous and heterologous prime-boost immunization strategies using novel and previously developed CHIKV vaccine candidates. Immunogenicity and efficacy were studied in a mouse model of CHIKV infection and showed that the DNA replicon and protein antigen were potent vaccine candidates, particularly when used for priming and boosting, respectively. Several prime-boost immunization strategies eliciting unmatched humoral and cellular immune responses were identified. Further characterization by antibody epitope mapping revealed differences in the qualitative immune responses induced by the different vaccine candidates and immunization strategies. Most vaccine modalities resulted in complete protection against wild-type CHIKV infection; however, we did identify circumstances under which certain immunization regimens may lead to enhancement of inflammation upon challenge. These results should help guide the design of CHIKV vaccine studies and will form the basis for further preclinical and clinical evaluation of these vaccine candidates. IMPORTANCE As of today, there is no licensed vaccine to prevent CHIKV infection. In considering potential new vaccine candidates, a vaccine that could raise long-term protective immunity after a single immunization would be preferable. While humoral immunity seems to be central for protection against CHIKV infection, we do not yet fully understand the correlates of protection. Therefore, in the absence of a functional vaccine, there is a need to evaluate a number of different candidates, assessing their merits when they are used either in a single immunization or in a homologous or heterologous prime-boost modality. Here we show that while single immunization with various vaccine candidates results in potent responses, combined approaches significantly enhance responses, suggesting that such approaches need to be considered in the further development of an efficacious CHIKV vaccine.
Collapse
|
9
|
Guo TC, Johansson DX, Haugland Ø, Liljeström P, Evensen Ø. A 6K-deletion variant of salmonid alphavirus is non-viable but can be rescued through RNA recombination. PLoS One 2014; 9:e100184. [PMID: 25009976 PMCID: PMC4091863 DOI: 10.1371/journal.pone.0100184] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2013] [Accepted: 05/23/2014] [Indexed: 01/13/2023] Open
Abstract
Pancreas disease (PD) of Atlantic salmon is an emerging disease caused by Salmonid alphavirus (SAV) which mainly affects salmonid aquaculture in Western Europe. Although genome structure of SAV has been characterized and each individual viral protein has been identified, the role of 6K protein in viral replication and infectivity remains undefined. The 6K protein of alphaviruses is a small and hydrophobic protein which is involved in membrane permeabilization, protein processing and virus budding. Because these common features are shared across many viral species, they have been named viroporins. In the present study, we applied reverse genetics to generate SAV3 6K-deleted (Δ6K) variant and investigate the role of 6K protein. Our findings show that the 6K-deletion variant of salmonid alphavirus is non-viable. Despite viral proteins of Δ6K variant are detected in the cytoplasm by immunostaining, they are not found on the cell surface. Further, analysis of viral proteins produced in Δ6K cDNA clone transfected cells using radioimmunoprecipitation (RIPA) and western blot showed a protein band of larger size than E2 of wild-type SAV3. When Δ6K cDNA was co-transfected with SAV3 helper cDNA encoding the whole structural genes including 6K, the infectivity was rescued. The development of CPE after co-transfection and resolved genome sequence of rescued virus confirmed full-length viral genome being generated through RNA recombination. The discovery of the important role of the 6K protein in virus production provides a new possibility for the development of antiviral intervention which is highly needed to control SAV infection in salmonids.
Collapse
Affiliation(s)
- Tz-Chun Guo
- Norwegian University of Life Sciences, Department of Basic Sciences and Aquatic Medicine, Oslo, Norway
| | - Daniel X. Johansson
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Øyvind Haugland
- Norwegian University of Life Sciences, Department of Basic Sciences and Aquatic Medicine, Oslo, Norway
| | - Peter Liljeström
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Øystein Evensen
- Norwegian University of Life Sciences, Department of Basic Sciences and Aquatic Medicine, Oslo, Norway
- * E-mail:
| |
Collapse
|
10
|
Wolf A, Hodneland K, Frost P, Hoeijmakers M, Rimstad E. Salmonid alphavirus-based replicon vaccine against infectious salmon anemia (ISA): impact of immunization route and interactions of the replicon vector. FISH & SHELLFISH IMMUNOLOGY 2014; 36:383-392. [PMID: 24374059 DOI: 10.1016/j.fsi.2013.12.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Revised: 11/26/2013] [Accepted: 12/18/2013] [Indexed: 06/03/2023]
Abstract
A salmonid alphavirus (SAV)-based replicon encoding the infectious salmon anemia virus (ISAV) hemagglutinin-esterase (HE), pSAV/HE, is an efficacious vaccine against infectious salmon anemia (ISA). Delivered intramuscularly (i.m.), the replicon vaccine provides high protection against subsequent ISAV challenge in Atlantic salmon (Salmo salar), and induces a strong innate response locally at the injection site. This may be beneficial and could warrant reduced doses and improved efficacy compared to conventional DNA vaccines. In the present study, we found that intraperitoneal (i.p.) administration of the pSAV/HE replicon vaccine did not induce protection, neither alone or in combination with a sub-potent, inactivated low-dose ISAV vaccine given i.p. No significant differences between the two immunization routes regarding systemic immune responses could be observed. I.m. injection of the replicon vector encoding a non-viral gene or the protective glycoprotein (G protein) from the heterologous viral hemorrhagic septicemia virus (VHSV) induced no protection against ISA. Although the replicons without the ISAV HE did induce IFN-signaling pathways at the muscle injection site similar to the pSAV/HE replicon they did not improve the efficacy of a sub-potent inactivated low-dose ISAV vaccine delivered i.p. Moreover, there was a tendency for reduced efficacy of the pSAV/HE replicon vaccine injected i.m. when co-injected with the replicon encoding the VHSV G protein, which previously, after DNA vaccination, have been reported to induce cross-protection against heterologous virus challenge in fish.
Collapse
Affiliation(s)
- Astrid Wolf
- Department of Food Safety and Infection Biology, The Norwegian School of Veterinary Science, P.O. 8146 Dep, N-0033 Oslo, Norway.
| | - Kjartan Hodneland
- MSD Animal Health Innovation AS, Thormøhlensgate 55, N-5008 Bergen, Norway.
| | - Petter Frost
- MSD Animal Health Innovation AS, Thormøhlensgate 55, N-5008 Bergen, Norway.
| | | | - Espen Rimstad
- Department of Food Safety and Infection Biology, The Norwegian School of Veterinary Science, P.O. 8146 Dep, N-0033 Oslo, Norway.
| |
Collapse
|
11
|
Aurisicchio L, Mancini R, Ciliberto G. Cancer vaccination by electro-gene-transfer. Expert Rev Vaccines 2014; 12:1127-37. [DOI: 10.1586/14760584.2013.836903] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
12
|
Knudsen ML, Ljungberg K, Liljeström P, Johansson DX. Intradermal electroporation of RNA. Methods Mol Biol 2014; 1121:147-54. [PMID: 24510820 DOI: 10.1007/978-1-4614-9632-8_13] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
This chapter describes the in vivo delivery of conventional mRNA or alphaviral replicon RNA via intradermal electroporation. The use of RNA in clinical applications has several potential advantages compared to DNA. For instance, RNA cannot integrate into the host genome, and it does not contain bacterial sequence motifs such as CpG often present in plasmid DNA backbones that can potentially trigger autoimmune responses. Intradermal electroporation is well tolerated and causes only minor trauma compared to intramuscular electroporation. As the skin houses high concentrations of antigen-presenting cells, vaccines could especially benefit from intradermal administration of RNA.
Collapse
Affiliation(s)
- Maria L Knudsen
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | | | | | | |
Collapse
|
13
|
Olsen CM, Pemula AK, Braaen S, Sankaran K, Rimstad E. Salmonid alphavirus replicon is functional in fish, mammalian and insect cells and in vivo in shrimps (Litopenaeus vannamei). Vaccine 2013; 31:5672-9. [DOI: 10.1016/j.vaccine.2013.09.058] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Revised: 09/18/2013] [Accepted: 09/24/2013] [Indexed: 10/26/2022]
|
14
|
Wolf A, Hodneland K, Frost P, Braaen S, Rimstad E. A hemagglutinin-esterase-expressing salmonid alphavirus replicon protects Atlantic salmon (Salmo salar) against infectious salmon anemia (ISA). Vaccine 2012. [PMID: 23200939 DOI: 10.1016/j.vaccine.2012.11.045] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
A replicon expression system based on the salmonid alphavirus (SAV) that encodes the infectious salmon anemia virus (ISAV) hemagglutinin-esterase (HE) was constructed and found to be an efficacious vaccine against infectious salmon anemia (ISA). Following a single intramuscular immunization, Atlantic salmon (Salmo salar) were effectively protected against subsequent ISAV challenge. Additional replicons coding for the ISAV fusion glycoprotein (F) or the ISAV matrix protein (M) were created and tested in combination with the replicon that encodes the HE. The ISAV HE was confirmed as a potent antigen, but neither the F nor the M proteins were found to be essential for immunization-induced protection. Innate immune response induced at the site of vaccination illustrated the immunogenicity of the SAV-based replicon and its ability to activate antiviral responses in Atlantic salmon. The successful testing of the SAV-based replicon as a vaccine model against ISA showed that the replicon approach may represent a novel immunization technology for the aquaculture industry. It offers potential benefits in terms of safety, efficacy, flexibility, and vaccine production complexity.
Collapse
Affiliation(s)
- Astrid Wolf
- Department of Food Safety and Infection Biology, The Norwegian School of Veterinary Science, N-0033 Oslo, Norway.
| | | | | | | | | |
Collapse
|
15
|
Yu YZ, Li N, Ma Y, Wang S, Yu WY, Sun ZW. Three types of human CpG motifs differentially modulate and augment immunogenicity of nonviral and viral replicon DNA vaccines as built-in adjuvants. Eur J Immunol 2012; 43:228-39. [PMID: 23037552 DOI: 10.1002/eji.201242690] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2012] [Revised: 09/14/2012] [Accepted: 10/01/2012] [Indexed: 01/20/2023]
Abstract
NakedDNA vaccines given by intramuscular injection are efficient in mouse models, but they require improvement for human use. As the immunogenicity of DNA vaccines depends, to a large extent, on the presence of CpG motifs as built-in adjuvants, we addressed this issue by inserting three types of human CpG motifs (A-type, B-type, and C-type) into the backbone of nonviral DNA and viral DNA replicon vectors with distinct immunostimulatory activities on human PBMCs. The adjuvant effects of CpG modifications in DNA vaccines expressing three types of antigens (β-Gal, AHc, or PA4) were then characterized in mice and found to significantly enhance antigen-specific humoral and cell-mediated immune responses. The three types of CpG motifs also differentially affected and modulated immune responses and protective potency against botulinum neurotoxin serotype A and Bacillus anthracis A16R challenge. Taken together, these results demonstrate that insertion of human CpG motifs can differentially modulate the immunogenicity of nonviral DNA vaccines as well as viral DNA replicon vaccines. Our study provides not only a better understanding of the in vivo activities of CpG motif adjuvants but implications for the rational design of such motifs as built-in adjuvants for DNA vectors targeting specific antigens.
Collapse
Affiliation(s)
- Yun-Zhou Yu
- Beijing Institute of Biotechnology, Beijing, China.
| | | | | | | | | | | |
Collapse
|
16
|
Superior induction of T cell responses to conserved HIV-1 regions by electroporated alphavirus replicon DNA compared to that with conventional plasmid DNA vaccine. J Virol 2012; 86:4082-90. [PMID: 22318135 DOI: 10.1128/jvi.06535-11] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Vaccination using "naked" DNA is a highly attractive strategy for induction of pathogen-specific immune responses; however, it has been only weakly immunogenic in humans. Previously, we constructed DNA-launched Semliki Forest virus replicons (DREP), which stimulate pattern recognition receptors and induce augmented immune responses. Also, in vivo electroporation was shown to enhance immune responses induced by conventional DNA vaccines. Here, we combine these two approaches and show that in vivo electroporation increases CD8(+) T cell responses induced by DREP and consequently decreases the DNA dose required to induce a response. The vaccines used in this study encode the multiclade HIV-1 T cell immunogen HIVconsv, which is currently being evaluated in clinical trials. Using intradermal delivery followed by electroporation, the DREP.HIVconsv DNA dose could be reduced to as low as 3.2 ng to elicit frequencies of HIV-1-specific CD8(+) T cells comparable to those induced by 1 μg of a conventional pTH.HIVconsv DNA vaccine, representing a 625-fold molar reduction in dose. Responses induced by both DREP.HIVconsv and pTH.HIVconsv were further increased by heterologous vaccine boosts employing modified vaccinia virus Ankara MVA.HIVconsv and attenuated chimpanzee adenovirus ChAdV63.HIVconsv. Using the same HIVconsv vaccines, the mouse observations were supported by an at least 20-fold-lower dose of DNA vaccine in rhesus macaques. These data point toward a strategy for overcoming the low immunogenicity of DNA vaccines in humans and strongly support further development of the DREP vaccine platform for clinical evaluation.
Collapse
|
17
|
Johansson DX, Ljungberg K, Kakoulidou M, Liljeström P. Intradermal electroporation of naked replicon RNA elicits strong immune responses. PLoS One 2012; 7:e29732. [PMID: 22238645 PMCID: PMC3251598 DOI: 10.1371/journal.pone.0029732] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2011] [Accepted: 12/04/2011] [Indexed: 11/18/2022] Open
Abstract
RNA-based vaccines represent an interesting immunization modality, but suffer from poor stability and a lack of efficient and clinically feasible delivery technologies. This study evaluates the immunogenic potential of naked in vitro transcribed Semliki Forest virus replicon RNA (RREP) delivered intradermally in combination with electroporation. Replicon-immunized mice showed a strong cellular and humoral response, contrary to mice immunized with regular mRNA. RREP-elicited induction of interferon-γ secreting CD8+ T cells and antibody responses were significantly increased by electroporation. CD8+ T cell responses remained substantial five weeks post vaccination, and antigen-specific CD8+ T cells with phenotypic characteristics of both effector and central memory cells were identified. The immune response during the contraction phase was further increased by a booster immunization, and the proportion of effector memory cells increased significantly. These results demonstrate that naked RREP delivered via intradermal electroporation constitute an immunogenic, safe and attractive alternative immunization strategy to DNA-based vaccines.
Collapse
Affiliation(s)
- Daniel X Johansson
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden.
| | | | | | | |
Collapse
|
18
|
Johansson DX, Ljungberg K, Kakoulidou M, Liljeström P. Intradermal electroporation of naked replicon RNA elicits strong immune responses. PLoS One 2012. [PMID: 22238645 DOI: 10.1371/journal.pone.0029732pone-d-11-11069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
RNA-based vaccines represent an interesting immunization modality, but suffer from poor stability and a lack of efficient and clinically feasible delivery technologies. This study evaluates the immunogenic potential of naked in vitro transcribed Semliki Forest virus replicon RNA (RREP) delivered intradermally in combination with electroporation. Replicon-immunized mice showed a strong cellular and humoral response, contrary to mice immunized with regular mRNA. RREP-elicited induction of interferon-γ secreting CD8+ T cells and antibody responses were significantly increased by electroporation. CD8+ T cell responses remained substantial five weeks post vaccination, and antigen-specific CD8+ T cells with phenotypic characteristics of both effector and central memory cells were identified. The immune response during the contraction phase was further increased by a booster immunization, and the proportion of effector memory cells increased significantly. These results demonstrate that naked RREP delivered via intradermal electroporation constitute an immunogenic, safe and attractive alternative immunization strategy to DNA-based vaccines.
Collapse
Affiliation(s)
- Daniel X Johansson
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden.
| | | | | | | |
Collapse
|
19
|
Sun Y, Li HY, Tian DY, Han QY, Zhang X, Li N, Qiu HJ. A novel alphavirus replicon-vectored vaccine delivered by adenovirus induces sterile immunity against classical swine fever. Vaccine 2011; 29:8364-72. [PMID: 21888938 DOI: 10.1016/j.vaccine.2011.08.085] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2011] [Revised: 08/15/2011] [Accepted: 08/16/2011] [Indexed: 11/28/2022]
Abstract
Low efficacy of gene-based vaccines due to inefficient gene delivery and expression has been major bottleneck of their applications. Efforts have been made to improve the efficacy, such as gene gun and electroporation, but the strategies are difficult to put into practical use. In this study, we developed and evaluated an adenovirus-delivered, alphavirus replicon-vectored vaccine (chimeric vector-based vaccine) expressing the E2 gene of classical swine fever virus (CSFV) (rAdV-SFV-E2). Rabbits immunized with rAdV-SFV-E2 developed CSFV-specific antibodies as early as 9 days and as long as 189 days and completely protected from challenge with C-strain. Pigs immunized with rAdV-SFV-E2 (n=5) developed robust humoral and cell-mediated responses to CSFV and were completely protected from subsequent lethal CSFV infection clinically and virologically. The level of immunity and protection induced by rAdV-SFV-E2 was comparable to that provided by the currently used live attenuated vaccine, C-strain. In contrast, both the conventional alphavirus replicon-vectored vaccine pSFV1CS-E2 and conventional adenovirus-vectored vaccine rAdV-E2 provided incomplete protection. The chimeric vector-based vaccine represents the first gene-based vaccine that is able to confer sterile immunity and complete protection against CSFV. The new-concept vaccination strategy may also be valuable in vaccine development against other pathogens.
Collapse
Affiliation(s)
- Yuan Sun
- Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | | | | | | | | | | | | |
Collapse
|
20
|
Näslund TI, Kostic L, Nordström EK, Chen M, Liljeström P. Role of innate signalling pathways in the immunogenicity of alphaviral replicon-based vaccines. Virol J 2011; 8:36. [PMID: 21261958 PMCID: PMC3038947 DOI: 10.1186/1743-422x-8-36] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2010] [Accepted: 01/24/2011] [Indexed: 11/10/2022] Open
Abstract
Background Alphaviral replicon-based vectors induce potent immune responses both when given as viral particles (VREP) or as DNA (DREP). It has been suggested that the strong immune stimulatory effect induced by these types of vectors is mediated by induction of danger signals and activation of innate signalling pathways due to the replicase activity. To investigate the innate signalling pathways involved, mice deficient in either toll-like receptors or downstream innate signalling molecules were immunized with DREP or VREP. Results We show that the induction of a CD8+ T cell response did not require functional TLR3 or MyD88 signalling. However, IRF3, converging several innate signalling pathways and important for generation of pro-inflammatory cytokines and type I IFNs, was needed for obtaining a robust primary immune response. Interestingly, type I interferon (IFN), induced by most innate signalling pathways, had a suppressing effect on both the primary and memory T cell responses after DREP and VREP immunization. Conclusions We show that alphaviral replicon-based vectors activate multiple innate signalling pathways, which both activate and restrict the induced immune response. These results further show that there is a delicate balance in the strength of innate signalling and induction of adaptive immune responses that should be taken into consideration when innate signalling molecules, such as type I IFNs, are used as vaccine adjuvant.
Collapse
Affiliation(s)
- Tanja I Näslund
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Nobels Väg 16, 17177 Stockholm, Sweden.
| | | | | | | | | |
Collapse
|
21
|
Distinct immune responses of recombinant plasmid DNA replicon vaccines expressing two types of antigens with or without signal sequences. Vaccine 2010; 28:7529-35. [DOI: 10.1016/j.vaccine.2010.08.107] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2010] [Revised: 07/12/2010] [Accepted: 08/13/2010] [Indexed: 11/19/2022]
|
22
|
Rosario M, Hopkins R, Fulkerson J, Borthwick N, Quigley MF, Joseph J, Douek DC, Greenaway HY, Venturi V, Gostick E, Price DA, Both GW, Sadoff JC, Hanke T. Novel recombinant Mycobacterium bovis BCG, ovine atadenovirus, and modified vaccinia virus Ankara vaccines combine to induce robust human immunodeficiency virus-specific CD4 and CD8 T-cell responses in rhesus macaques. J Virol 2010; 84:5898-908. [PMID: 20375158 PMCID: PMC2876636 DOI: 10.1128/jvi.02607-09] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2009] [Accepted: 03/30/2010] [Indexed: 11/20/2022] Open
Abstract
Mycobacterium bovis bacillus Calmette-Guérin (BCG), which elicits a degree of protective immunity against tuberculosis, is the most widely used vaccine in the world. Due to its persistence and immunogenicity, BCG has been proposed as a vector for vaccines against other infections, including HIV-1. BCG has a very good safety record, although it can cause disseminated disease in immunocompromised individuals. Here, we constructed a recombinant BCG vector expressing HIV-1 clade A-derived immunogen HIVA using the recently described safer and more immunogenic BCG strain AERAS-401 as the parental mycobacterium. Using routine ex vivo T-cell assays, BCG.HIVA(401) as a stand-alone vaccine induced undetectable and weak CD8 T-cell responses in BALB/c mice and rhesus macaques, respectively. However, when BCG.HIVA(401) was used as a priming component in heterologous vaccination regimens together with recombinant modified vaccinia virus Ankara-vectored MVA.HIVA and ovine atadenovirus-vectored OAdV.HIVA vaccines, robust HIV-1-specific T-cell responses were elicited. These high-frequency T-cell responses were broadly directed and capable of proliferation in response to recall antigen. Furthermore, multiple antigen-specific T-cell clonotypes were efficiently recruited into the memory pool. These desirable features are thought to be associated with good control of HIV-1 infection. In addition, strong and persistent T-cell responses specific for the BCG-derived purified protein derivative (PPD) antigen were induced. This work is the first demonstration of immunogenicity for two novel vaccine vectors and the corresponding candidate HIV-1 vaccines BCG.HIVA(401) and OAdV.HIVA in nonhuman primates. These results strongly support their further exploration.
Collapse
Affiliation(s)
- Maximillian Rosario
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe, Oxford OX3 9DS, United Kingdom, Aeras Global TB Vaccine Foundation, 1405 Research Blvd., Rockville, Maryland 20850, Vaccine Research Centre, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, Catalan HIV Vaccine Research and Development Center, AIDS Research Unit, Infectious Diseases Department, Hospital Clinic, August Pi i Sunyer Biomedical Research Institute, School of Medicine, University of Barcelona, 170 08036 Barcelona, Spain, Computational Biology Unit, Centre for Vascular Research, University of New South Wales, Kensington, New South Wales 2052, Australia, Department of Medical Biochemistry and Immunology, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom, Biotech Equity Partners Pty., Ltd., Riverside Life Sciences Building, 11 Julius Ave., North Ryde, New South Wales 2113, Australia
| | - Richard Hopkins
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe, Oxford OX3 9DS, United Kingdom, Aeras Global TB Vaccine Foundation, 1405 Research Blvd., Rockville, Maryland 20850, Vaccine Research Centre, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, Catalan HIV Vaccine Research and Development Center, AIDS Research Unit, Infectious Diseases Department, Hospital Clinic, August Pi i Sunyer Biomedical Research Institute, School of Medicine, University of Barcelona, 170 08036 Barcelona, Spain, Computational Biology Unit, Centre for Vascular Research, University of New South Wales, Kensington, New South Wales 2052, Australia, Department of Medical Biochemistry and Immunology, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom, Biotech Equity Partners Pty., Ltd., Riverside Life Sciences Building, 11 Julius Ave., North Ryde, New South Wales 2113, Australia
| | - John Fulkerson
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe, Oxford OX3 9DS, United Kingdom, Aeras Global TB Vaccine Foundation, 1405 Research Blvd., Rockville, Maryland 20850, Vaccine Research Centre, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, Catalan HIV Vaccine Research and Development Center, AIDS Research Unit, Infectious Diseases Department, Hospital Clinic, August Pi i Sunyer Biomedical Research Institute, School of Medicine, University of Barcelona, 170 08036 Barcelona, Spain, Computational Biology Unit, Centre for Vascular Research, University of New South Wales, Kensington, New South Wales 2052, Australia, Department of Medical Biochemistry and Immunology, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom, Biotech Equity Partners Pty., Ltd., Riverside Life Sciences Building, 11 Julius Ave., North Ryde, New South Wales 2113, Australia
| | - Nicola Borthwick
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe, Oxford OX3 9DS, United Kingdom, Aeras Global TB Vaccine Foundation, 1405 Research Blvd., Rockville, Maryland 20850, Vaccine Research Centre, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, Catalan HIV Vaccine Research and Development Center, AIDS Research Unit, Infectious Diseases Department, Hospital Clinic, August Pi i Sunyer Biomedical Research Institute, School of Medicine, University of Barcelona, 170 08036 Barcelona, Spain, Computational Biology Unit, Centre for Vascular Research, University of New South Wales, Kensington, New South Wales 2052, Australia, Department of Medical Biochemistry and Immunology, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom, Biotech Equity Partners Pty., Ltd., Riverside Life Sciences Building, 11 Julius Ave., North Ryde, New South Wales 2113, Australia
| | - Máire F. Quigley
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe, Oxford OX3 9DS, United Kingdom, Aeras Global TB Vaccine Foundation, 1405 Research Blvd., Rockville, Maryland 20850, Vaccine Research Centre, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, Catalan HIV Vaccine Research and Development Center, AIDS Research Unit, Infectious Diseases Department, Hospital Clinic, August Pi i Sunyer Biomedical Research Institute, School of Medicine, University of Barcelona, 170 08036 Barcelona, Spain, Computational Biology Unit, Centre for Vascular Research, University of New South Wales, Kensington, New South Wales 2052, Australia, Department of Medical Biochemistry and Immunology, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom, Biotech Equity Partners Pty., Ltd., Riverside Life Sciences Building, 11 Julius Ave., North Ryde, New South Wales 2113, Australia
| | - Joan Joseph
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe, Oxford OX3 9DS, United Kingdom, Aeras Global TB Vaccine Foundation, 1405 Research Blvd., Rockville, Maryland 20850, Vaccine Research Centre, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, Catalan HIV Vaccine Research and Development Center, AIDS Research Unit, Infectious Diseases Department, Hospital Clinic, August Pi i Sunyer Biomedical Research Institute, School of Medicine, University of Barcelona, 170 08036 Barcelona, Spain, Computational Biology Unit, Centre for Vascular Research, University of New South Wales, Kensington, New South Wales 2052, Australia, Department of Medical Biochemistry and Immunology, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom, Biotech Equity Partners Pty., Ltd., Riverside Life Sciences Building, 11 Julius Ave., North Ryde, New South Wales 2113, Australia
| | - Daniel C. Douek
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe, Oxford OX3 9DS, United Kingdom, Aeras Global TB Vaccine Foundation, 1405 Research Blvd., Rockville, Maryland 20850, Vaccine Research Centre, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, Catalan HIV Vaccine Research and Development Center, AIDS Research Unit, Infectious Diseases Department, Hospital Clinic, August Pi i Sunyer Biomedical Research Institute, School of Medicine, University of Barcelona, 170 08036 Barcelona, Spain, Computational Biology Unit, Centre for Vascular Research, University of New South Wales, Kensington, New South Wales 2052, Australia, Department of Medical Biochemistry and Immunology, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom, Biotech Equity Partners Pty., Ltd., Riverside Life Sciences Building, 11 Julius Ave., North Ryde, New South Wales 2113, Australia
| | - Hui Yee Greenaway
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe, Oxford OX3 9DS, United Kingdom, Aeras Global TB Vaccine Foundation, 1405 Research Blvd., Rockville, Maryland 20850, Vaccine Research Centre, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, Catalan HIV Vaccine Research and Development Center, AIDS Research Unit, Infectious Diseases Department, Hospital Clinic, August Pi i Sunyer Biomedical Research Institute, School of Medicine, University of Barcelona, 170 08036 Barcelona, Spain, Computational Biology Unit, Centre for Vascular Research, University of New South Wales, Kensington, New South Wales 2052, Australia, Department of Medical Biochemistry and Immunology, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom, Biotech Equity Partners Pty., Ltd., Riverside Life Sciences Building, 11 Julius Ave., North Ryde, New South Wales 2113, Australia
| | - Vanessa Venturi
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe, Oxford OX3 9DS, United Kingdom, Aeras Global TB Vaccine Foundation, 1405 Research Blvd., Rockville, Maryland 20850, Vaccine Research Centre, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, Catalan HIV Vaccine Research and Development Center, AIDS Research Unit, Infectious Diseases Department, Hospital Clinic, August Pi i Sunyer Biomedical Research Institute, School of Medicine, University of Barcelona, 170 08036 Barcelona, Spain, Computational Biology Unit, Centre for Vascular Research, University of New South Wales, Kensington, New South Wales 2052, Australia, Department of Medical Biochemistry and Immunology, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom, Biotech Equity Partners Pty., Ltd., Riverside Life Sciences Building, 11 Julius Ave., North Ryde, New South Wales 2113, Australia
| | - Emma Gostick
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe, Oxford OX3 9DS, United Kingdom, Aeras Global TB Vaccine Foundation, 1405 Research Blvd., Rockville, Maryland 20850, Vaccine Research Centre, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, Catalan HIV Vaccine Research and Development Center, AIDS Research Unit, Infectious Diseases Department, Hospital Clinic, August Pi i Sunyer Biomedical Research Institute, School of Medicine, University of Barcelona, 170 08036 Barcelona, Spain, Computational Biology Unit, Centre for Vascular Research, University of New South Wales, Kensington, New South Wales 2052, Australia, Department of Medical Biochemistry and Immunology, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom, Biotech Equity Partners Pty., Ltd., Riverside Life Sciences Building, 11 Julius Ave., North Ryde, New South Wales 2113, Australia
| | - David A. Price
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe, Oxford OX3 9DS, United Kingdom, Aeras Global TB Vaccine Foundation, 1405 Research Blvd., Rockville, Maryland 20850, Vaccine Research Centre, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, Catalan HIV Vaccine Research and Development Center, AIDS Research Unit, Infectious Diseases Department, Hospital Clinic, August Pi i Sunyer Biomedical Research Institute, School of Medicine, University of Barcelona, 170 08036 Barcelona, Spain, Computational Biology Unit, Centre for Vascular Research, University of New South Wales, Kensington, New South Wales 2052, Australia, Department of Medical Biochemistry and Immunology, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom, Biotech Equity Partners Pty., Ltd., Riverside Life Sciences Building, 11 Julius Ave., North Ryde, New South Wales 2113, Australia
| | - Gerald W. Both
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe, Oxford OX3 9DS, United Kingdom, Aeras Global TB Vaccine Foundation, 1405 Research Blvd., Rockville, Maryland 20850, Vaccine Research Centre, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, Catalan HIV Vaccine Research and Development Center, AIDS Research Unit, Infectious Diseases Department, Hospital Clinic, August Pi i Sunyer Biomedical Research Institute, School of Medicine, University of Barcelona, 170 08036 Barcelona, Spain, Computational Biology Unit, Centre for Vascular Research, University of New South Wales, Kensington, New South Wales 2052, Australia, Department of Medical Biochemistry and Immunology, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom, Biotech Equity Partners Pty., Ltd., Riverside Life Sciences Building, 11 Julius Ave., North Ryde, New South Wales 2113, Australia
| | - Jerald C. Sadoff
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe, Oxford OX3 9DS, United Kingdom, Aeras Global TB Vaccine Foundation, 1405 Research Blvd., Rockville, Maryland 20850, Vaccine Research Centre, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, Catalan HIV Vaccine Research and Development Center, AIDS Research Unit, Infectious Diseases Department, Hospital Clinic, August Pi i Sunyer Biomedical Research Institute, School of Medicine, University of Barcelona, 170 08036 Barcelona, Spain, Computational Biology Unit, Centre for Vascular Research, University of New South Wales, Kensington, New South Wales 2052, Australia, Department of Medical Biochemistry and Immunology, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom, Biotech Equity Partners Pty., Ltd., Riverside Life Sciences Building, 11 Julius Ave., North Ryde, New South Wales 2113, Australia
| | - Tomáš Hanke
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe, Oxford OX3 9DS, United Kingdom, Aeras Global TB Vaccine Foundation, 1405 Research Blvd., Rockville, Maryland 20850, Vaccine Research Centre, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, Catalan HIV Vaccine Research and Development Center, AIDS Research Unit, Infectious Diseases Department, Hospital Clinic, August Pi i Sunyer Biomedical Research Institute, School of Medicine, University of Barcelona, 170 08036 Barcelona, Spain, Computational Biology Unit, Centre for Vascular Research, University of New South Wales, Kensington, New South Wales 2052, Australia, Department of Medical Biochemistry and Immunology, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom, Biotech Equity Partners Pty., Ltd., Riverside Life Sciences Building, 11 Julius Ave., North Ryde, New South Wales 2113, Australia
| |
Collapse
|
23
|
Pan Y, Zhao Q, Fang L, Luo R, Chen H, Xiao S. Efficient gene delivery into mammalian cells by recombinant baculovirus containing a hybrid cytomegalovirus promoter/Semliki Forest virus replicon. J Gene Med 2010; 11:1030-8. [PMID: 19757479 DOI: 10.1002/jgm.1390] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Baculovirus, which is widely utilized as an excellent tool for the production of recombinant protein in insect cells, has recently emerged as a novel and attractive gene delivery vehicle for mammalian cells. Alphavirus, such as Semliki Forest virus (SFV), has also received considerable attention for use as expression vectors because of its self-replicating property. In the present study, we investigated the characterization of recombinant baculovirus incorporating a hybrid cytomegalovirus (CMV) promoter/SFV replicon. METHODS Recombinant baculovirus containing the hybrid CMV promoter/SFV replicon was constructed. Using enhanced green fluorescence protein (EGFP) as the reporter gene, gene delivery efficiencies and the ability to express heterogenous protein in mammalian cells were evaluated. Optimal transduction conditions, including transduction temperature, time and dose, were also investigated. RESULTS The obtained recombinant baculovirus, Bac-CMV/SFV-EGFP, exhibited high transduction efficiency and high-level expression of reporter protein in mammalian cells. Furthermore, this recombinant baculovirus could induce apoptosis in mammalian cells in the course of transduction, as demonstrated by the observed DNA laddering patterns and increased caspase-3 activity. CONCLUSIONS The developed baculovirus vector has a high transduction efficiency and the ability to mediate foreign gene expression in mammalian cells. Taken together with its pro-apoptotic properties, this baculovirus vector may provide an alternative tool for vaccine development.
Collapse
Affiliation(s)
- Yongfei Pan
- Division of Animal Infectious Diseases, State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | | | | | | | | | | |
Collapse
|
24
|
Yu Y, Yu J, Li N, Wang S, Yu W, Sun Z. Individual and bivalent vaccines against botulinum neurotoxin serotypes A and B using DNA-based Semliki Forest virus vectors. Vaccine 2009; 27:6148-53. [DOI: 10.1016/j.vaccine.2009.08.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2009] [Revised: 07/29/2009] [Accepted: 08/06/2009] [Indexed: 11/29/2022]
|
25
|
Saxena S, Dahiya SS, Sonwane AA, Patel CL, Saini M, Rai A, Gupta PK. A sindbis virus replicon-based DNA vaccine encoding the rabies virus glycoprotein elicits immune responses and complete protection in mice from lethal challenge. Vaccine 2009; 26:6592-601. [PMID: 18848857 DOI: 10.1016/j.vaccine.2008.09.055] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2008] [Revised: 09/14/2008] [Accepted: 09/15/2008] [Indexed: 11/17/2022]
Abstract
A sindbis virus replicon-based DNA vaccine encoding rabies virus glycoprotein (G) was developed by subcloning rabies G gene into a sindbis virus replicon-based vaccine vector (pAlpha). The self-amplification of RNA transcripts and translation efficiency of rabies G was analyzed in pAlpha-Rab-G-transfected mammalian cells using RT-PCR, SDS-PAGE and Western blot analysis. The transfected cells also showed induction of apoptosis which is an important event in the enhancement of immune responses. Further, immune responses induced with replicon-based rabies DNA vaccine (pAlpha-Rab-G) was compared with conventional rabies DNA vaccine and commercial cell culture vaccine (Rabipur) in intramuscularly injected mice. The mice immunized with replicon-based rabies DNA vaccine induced humoral and cell mediated immune responses better than conventional rabies DNA vaccine however, comparable to Rabipur vaccine. On challenge with rabies virus CVS strain, replicon-based rabies DNA vaccine conferred complete protection similar to Rabipur. These results demonstrate that replicon-based rabies DNA vaccine is effective in inducing both humoral and cellular immune responses and can be considered as effective vaccine against rabies.
Collapse
Affiliation(s)
- Sonal Saxena
- Division of Animal Biotechnology, Indian Veterinary Research Institute, Izatnagar 243122, India
| | | | | | | | | | | | | |
Collapse
|
26
|
Induction of immune responses and protection in mice against rabies using a self-replicating RNA vaccine encoding rabies virus glycoprotein. Vet Microbiol 2009; 136:36-44. [DOI: 10.1016/j.vetmic.2008.10.030] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2008] [Revised: 10/19/2008] [Accepted: 10/28/2008] [Indexed: 11/23/2022]
|
27
|
Rausalu K, Iofik A, Ulper L, Karo-Astover L, Lulla V, Merits A. Properties and use of novel replication-competent vectors based on Semliki Forest virus. Virol J 2009; 6:33. [PMID: 19317912 PMCID: PMC2669057 DOI: 10.1186/1743-422x-6-33] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2009] [Accepted: 03/24/2009] [Indexed: 12/27/2022] Open
Abstract
Background Semliki Forest virus (SFV) has a positive strand RNA genome and infects different cells of vertebrates and invertebrates. The 5' two-thirds of the genome encodes non-structural proteins that are required for virus replication and synthesis of subgenomic (SG) mRNA for structural proteins. SG-mRNA is generated by internal initiation at the SG-promoter that is located at the complementary minus-strand template. Different types of expression systems including replication-competent vectors, which represent alphavirus genomes with inserted expression units, have been developed. The replication-competent vectors represent useful tools for studying alphaviruses and have potential therapeutic applications. In both cases, the properties of the vector, such as its genetic stability and expression level of the protein of interest, are important. Results We analysed 14 candidates of replication-competent vectors based on the genome of an SFV4 isolate that contained a duplicated SG promoter or an internal ribosomal entry site (IRES)-element controlled marker gene. It was found that the IRES elements and the minimal -21 to +5 SG promoter were non-functional in the context of these vectors. The efficient SG promoters contained at least 26 residues upstream of the start site of SG mRNA. The insertion site of the SG promoter and its length affected the genetic stability of the vectors, which was always higher when the SG promoter was inserted downstream of the coding region for structural proteins. The stability also depended on the conditions used for vector propagation. A procedure based on the in vitro transcription of ligation products was used for generation of replication-competent vector-based expression libraries that contained hundreds of thousands of different genomes, and maintained genetic diversity and the ability to express inserted genes over five passages in cell culture. Conclusion The properties of replication-competent vectors of alphaviruses depend on the details of their construction. In the case of SFV4, such vectors should contain the SG promoter with structural characteristics for this isolate. The main factor for instability of SFV4-based replication-competent vectors was the deletion of genes of interest, since the resulting shorter genomes had a growth advantage over the original vector.
Collapse
Affiliation(s)
- Kai Rausalu
- Institute of Technology, University of Tartu, Nooruse 1, 50411, Tartu, Estonia.
| | | | | | | | | | | |
Collapse
|
28
|
Zhao HP, Sun JF, Li N, Sun Y, Xia ZH, Wang Y, Cheng D, Qi QF, Jin ML, Qiu HJ. Assessment of the cell-mediated immunity induced by alphavirus replicon-vectored DNA vaccines against classical swine fever in a mouse model. Vet Immunol Immunopathol 2008; 129:57-65. [PMID: 19150136 DOI: 10.1016/j.vetimm.2008.12.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2008] [Revised: 12/01/2008] [Accepted: 12/04/2008] [Indexed: 11/30/2022]
Abstract
We have previously shown that an alphavirus replicon-vectored DNA vaccine (pSFV1CS-E2) encoding the E2 glycoprotein of classical swine fever virus (CSFV) completely protected the immunized pigs from lethal challenge. These animals developed only low or moderate level viral-specific antibody titers before challenge, implying that cell-mediated immunity (CMI) probably played an important role in the protective immunity against CSFV conferred by the DNA vaccine. In this study, the CMI induced by pSFV1CS-E2 and its derivative pSFV1CS-E2-UL49 encoding a fusion protein of CSFV E2 and pseudorabies virus (PRV) VP22 was evaluated in a mouse model by lymphoproliferation assays based on CFSE or WST-8, intracellular cytokine staining, and cytokine ELISA. The results showed that both vaccines induced CSFV-specific lymphoproliferative responses and cytokine production, and pSFV1CS-E2-UL49 induced stronger lymphoproliferative responses and higher cytokine levels than pSFV1CS-E2. These findings suggest that the alphavirus replicon-delivered DNA vaccines are capable of inducing CMI, and PRV VP22 is able to enhance the immunogenicity of the co-delivered antigen.
Collapse
Affiliation(s)
- He-Ping Zhao
- Division of Swine Infectious Diseases, National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin, Heilongjiang, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Human immunodeficiency virus type 1 env trimer immunization of macaques and impact of priming with viral vector or stabilized core protein. J Virol 2008; 83:540-51. [PMID: 19004960 DOI: 10.1128/jvi.01102-08] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Currently there is limited information about the quality of immune responses elicited by candidate human immunodeficiency virus type 1 (HIV-1) envelope glycoprotein (Env)-based immunogens in primates. Here we describe a comprehensive analysis of neutralizing antibody and T-cell responses obtained in cynomolgus macaques by three selected immunization regimens. We used the previously described YU2-based gp140 protein trimers administered in an adjuvant, preceded by two distinct priming strategies: either alphavirus replicon particles expressing matched gp140 trimers or gp120 core proteins stabilized in the CD4-bound conformation. The rationale for priming with replicon particles was to evaluate the impact of the expression platform on trimer immunogenicity. The stable core proteins were chosen in an attempt to expand selectively lymphocytes recognizing common determinants between the core and trimers to broaden the immune response. The results presented here demonstrate that the platform by which Env trimers were delivered in the priming (either protein or replicon vector) had little impact on the overall immune response. In contrast, priming with stable core proteins followed by a trimer boost strikingly focused the T-cell response on the core sequences of HIV-1 Env. The specificity of the T-cell response was distinctly different from that of the responses obtained in animals immunized with trimers alone and was shown to be mediated by CD4(+) T cells. However, this regimen showed limited or no improvement in the neutralizing antibody responses, suggesting that further immunogen design efforts are required to successfully focus the B-cell response on conserved neutralizing determinants of HIV-1 Env.
Collapse
|
30
|
Construction, properties, and potential application of infectious plasmids containing Semliki Forest virus full-length cDNA with an inserted intron. J Virol Methods 2007; 148:265-70. [PMID: 18054090 PMCID: PMC7172237 DOI: 10.1016/j.jviromet.2007.10.007] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2007] [Revised: 10/04/2007] [Accepted: 10/10/2007] [Indexed: 12/03/2022]
Abstract
Semliki Forest virus (SFV, genus Alphavirus) has a broad host range, high efficiency of viral protein expression, and the ability to stimulate an immune response. These properties have made SFV an attractive tool for development of expression vectors, and plasmid clones containing cDNA of the SFV genome often are used. However, instability of these plasmids resulting from cryptic expression of SFV envelope proteins in Escherichia coli represents a problem both for the development of SFV-based vectors and for SFV research. In this study, an infectious plasmid of SFV, pCMV-SFV4, was constructed; its toxic effect was eliminated by intron insertion in the capsid protein encoding region. When transfected into mammalian cells, the plasmid clone was highly infectious and produced virus with properties identical to those of wild-type SFV. The inserted intron was efficiently and properly removed from the RNA genome of SFV. Therefore, this novel and stabilized infectious SFV plasmid represents a superior tool for basic studies of SFV as well as for biotechnological applications.
Collapse
|
31
|
Yu YZ, Zhang SM, Sun ZW, Wang S, Yu WY. Enhanced immune responses using plasmid DNA replicon vaccine encoding the Hc domain of Clostridium botulinum neurotoxin serotype A. Vaccine 2007; 25:8843-50. [PMID: 18022294 DOI: 10.1016/j.vaccine.2007.10.016] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2007] [Revised: 09/14/2007] [Accepted: 10/07/2007] [Indexed: 11/19/2022]
Abstract
In current study, the immunogenicity of a plasmid DNA replicon vaccine (pSCARSHc) encoding the Hc domain of Clostridium botulinum neurotoxin serotype A (AHc) was investigated and compared with a conventional plasmid DNA vaccine (pcDNASHc) encoding the same antigen. In vitro, pSCARSHc incorporating Semliki Forest virus (SFV) replicon could express AHc protein and induce apoptosis of transfected cells. Comparison with the conventional plasmid DNA vaccine (pcDNASHc) yielded several interesting results. First, our self-designed pSCARSHc could induce relatively higher AHc-specific antibodies and lymphocyte proliferative responses in immunized Balb/c mice, especially at low doses. Second, while both pSCARSHc and pcDNASHc induced Th2-type immune responses, the ratio of IgG1 to IgG2a was lower in pSCARSHc groups and the Th2- and Th1-type humoral immune responses induced by pSCARSHc were also stronger than that of the pcDNASHc vaccine. Third, it was shown that the sera from pSCARSHc-vaccinated mice conferred more efficient protection than those from pcDNASHc-vaccinated mice by BoNT/A neutralization assay. Finally, mice immunized with pSCARSHc could also elicit more efficient protection against BoNT/A than pcDNASHc. These results indicate that our plasmid DNA replicon vaccine can provide strong immunogenicity and should be a potential alternative strategy to conventional DNA vaccines in developing an efficacious vaccine against C. botulinum neurotoxin serotype A.
Collapse
Affiliation(s)
- Yun-Zhou Yu
- Beijing Institute of Biotechnology, 20 Dong Dajie Street, Fengtai District, Beijing 100071, China
| | | | | | | | | |
Collapse
|
32
|
Liu MA, Wahren B, Karlsson Hedestam GB. DNA vaccines: recent developments and future possibilities. Hum Gene Ther 2007; 17:1051-61. [PMID: 17032152 DOI: 10.1089/hum.2006.17.1051] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The field of DNA vaccines continues to advance and several new strategies to augment the immunogenicity of DNA vaccines are under evaluation. The majority of these studies are in the early preclinical stage, but some DNA vaccines have moved into clinical trials. In this review, we describe some of the more recent efforts aimed at increasing the immunogenicity of DNA vaccines, including the use of genetic adjuvants and plasmid-based expression of viral replicons. In addition, we discuss the possibility of using DNA vaccines to address emerging infectious agents where they may provide an advantage over other vaccine strategies and we review some areas where DNA vaccines have been used to target self-antigens.
Collapse
|
33
|
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
|
34
|
Liu MA, Wahren B, Hedestam GBK. DNA Vaccines: Recent Developments and Future Possibilities. Hum Gene Ther 2006. [DOI: 10.1089/hum.2006.17.ft-250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
35
|
Li S, Locke E, Bruder J, Clarke D, Doolan DL, Havenga MJE, Hill AVS, Liljestrom P, Monath TP, Naim HY, Ockenhouse C, Tang DCC, Van Kampen KR, Viret JF, Zavala F, Dubovsky F. Viral vectors for malaria vaccine development. Vaccine 2006; 25:2567-74. [PMID: 16914237 PMCID: PMC7131149 DOI: 10.1016/j.vaccine.2006.07.035] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2006] [Revised: 06/23/2006] [Accepted: 07/23/2006] [Indexed: 01/08/2023]
Abstract
A workshop on viral vectors for malaria vaccine development, organized by the PATH Malaria Vaccine Initiative, was held in Bethesda, MD on October 20, 2005. Recent advancements in viral-vectored malaria vaccine development and emerging vector technologies were presented and discussed. Classic viral vectors such as poxvirus, adenovirus and alphavirus vectors have been successfully used to deliver malaria antigens. Some of the vaccine candidates have demonstrated their potential in inducing malaria-specific immunity in animal models and human trials. In addition, emerging viral-vector technologies, such as measles virus (MV), vesicular stomatitis virus (VSV) and yellow fever (YF) virus, may also be useful for malaria vaccine development. Studies in animal models suggest that each viral vector is unique in its ability to induce humoral and/or cellular immune responses. Those studies have also revealed that optimization of Plasmodium genes for mammalian expression is an important aspect of vaccine design. Codon-optimization, surface-trafficking, de-glycosylation and removal of toxic domains can lead to improved immunogenicity. Understanding the vector's ability to induce an immune response and the expression of malaria antigens in mammalian cells will be critical in designing the next generation of viral-vectored malaria vaccines.
Collapse
|
36
|
Riezebos-Brilman A, de Mare A, Bungener L, Huckriede A, Wilschut J, Daemen T. Recombinant alphaviruses as vectors for anti-tumour and anti-microbial immunotherapy. J Clin Virol 2006; 35:233-43. [PMID: 16448844 DOI: 10.1016/j.jcv.2005.12.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2005] [Accepted: 12/07/2005] [Indexed: 11/15/2022]
Abstract
BACKGROUND Vectors derived from alphaviruses are gaining interest for their high transfection potency and strong immunogenicity. OBJECTIVES After a brief introduction on alphaviruses and their vectors, an overview is given on current preclinical immunotherapy studies using vector systems based on alphaviruses. The efficacy of alphavirus vectors in inducing immune responses will be illustrated by a more detailed description of immunization studies using recombinant Semliki Forest virus for the treatment of human papilloma virus-induced cervical cancer. RESULTS Immunization with recombinant alphavirus results in the induction of humoral and cellular immune responses against microbes, infected cells and cancer cells. Preclinical studies demonstrate that infectious diseases and cancer can be treated prophylactically as well as therapeutically. CONCLUSIONS Alphavirus-based genetic immunization strategies are highly effective in animal model systems, comparing quite favourably with any other approach. Therefore, we hope and expect to see an efficient induction of tumour-or microbial immunity and a positive outcome in future clinical efficacy studies.
Collapse
Affiliation(s)
- Annelies Riezebos-Brilman
- Department of Medical Microbiology, Molecular Virology Section, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | | | | | | | | | | |
Collapse
|