1
|
|
2
|
Abstract
Many nonhuman adenoviruses (AdVs) of simian, bovine, porcine, canine, ovine, murine, and fowl origin are being developed as gene delivery systems for recombinant vaccines and gene therapy applications. In addition to circumventing preexisting human AdV (HAdV) immunity, nonhuman AdV vectors utilize coxsackievirus-adenovirus receptor or other receptors for vector internalization, thereby expanding the range of cell types that can be targeted. Nonhuman AdV vectors also provide excellent platforms for veterinary vaccines. A specific nonhuman AdV vector when used in its species of origin could provide an excellent animal model for evaluating the vector efficacy and pathogenesis. These vectors are useful in prime–boost approaches with other AdV vectors or with other gene delivery systems including DNA immunization and viral or bacterial vectors. When multiple vector inoculations are required, nonhuman AdV vectors could supplement HAdV or other viral vectors.
Collapse
|
3
|
Uusi-Kerttula H, Hulin-Curtis S, Davies J, Parker AL. Oncolytic Adenovirus: Strategies and Insights for Vector Design and Immuno-Oncolytic Applications. Viruses 2015; 7:6009-42. [PMID: 26610547 PMCID: PMC4664994 DOI: 10.3390/v7112923] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 11/13/2015] [Accepted: 11/17/2015] [Indexed: 02/06/2023] Open
Abstract
Adenoviruses (Ad) are commonly used both experimentally and clinically, including oncolytic virotherapy applications. In the clinical area, efficacy is frequently hampered by the high rates of neutralizing immunity, estimated as high as 90% in some populations that promote vector clearance and limit bioavailability for tumor targeting following systemic delivery. Active tumor targeting is also hampered by the ubiquitous nature of the Ad5 receptor, hCAR, as well as the lack of highly tumor-selective targeting ligands and suitable targeting strategies. Furthermore, significant off-target interactions between the viral vector and cellular and proteinaceous components of the bloodstream have been documented that promote uptake into non-target cells and determine dose-limiting toxicities. Novel strategies are therefore needed to overcome the obstacles that prevent efficacious Ad deployment for wider clinical applications. The use of less seroprevalent Ad serotypes, non-human serotypes, capsid pseudotyping, chemical shielding and genetic masking by heterologous peptide incorporation are all potential strategies to achieve efficient vector escape from humoral immune recognition. Conversely, selective vector arming with immunostimulatory agents can be utilized to enhance their oncolytic potential by activation of cancer-specific immune responses against the malignant tissues. This review presents recent advantages and pitfalls occurring in the field of adenoviral oncolytic therapies.
Collapse
Affiliation(s)
- Hanni Uusi-Kerttula
- Institute of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff CF14 4XN, UK.
| | - Sarah Hulin-Curtis
- Institute of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff CF14 4XN, UK.
| | - James Davies
- Institute of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff CF14 4XN, UK.
| | - Alan L Parker
- Institute of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff CF14 4XN, UK.
| |
Collapse
|
4
|
Development of Novel Adenoviral Vectors to Overcome Challenges Observed With HAdV-5-based Constructs. Mol Ther 2015; 24:6-16. [PMID: 26478249 PMCID: PMC4754553 DOI: 10.1038/mt.2015.194] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 10/07/2015] [Indexed: 12/23/2022] Open
Abstract
Recombinant vectors based on human adenovirus serotype 5 (HAdV-5) have been extensively studied in preclinical models and clinical trials over the past two decades. However, the thorough understanding of the HAdV-5 interaction with human subjects has uncovered major concerns about its product applicability. High vector-associated toxicity and widespread preexisting immunity have been shown to significantly impede the effectiveness of HAdV-5–mediated gene transfer. It is therefore that the in-depth knowledge attained working on HAdV-5 is currently being used to develop alternative vectors. Here, we provide a comprehensive overview of data obtained in recent years disqualifying the HAdV-5 vector for systemic gene delivery as well as novel strategies being pursued to overcome the limitations observed with particular emphasis on the ongoing vectorization efforts to obtain vectors based on alternative serotypes.
Collapse
|
5
|
Fausther-Bovendo H, Kobinger GP. Pre-existing immunity against Ad vectors: humoral, cellular, and innate response, what's important? Hum Vaccin Immunother 2015; 10:2875-84. [PMID: 25483662 PMCID: PMC5443060 DOI: 10.4161/hv.29594] [Citation(s) in RCA: 180] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Pre-existing immunity against human adenovirus (HAd) serotype 5 derived vector in the human population is widespread, thus hampering its clinical use. Various components of the immune system, including neutralizing antibodies (nAbs), Ad specific T cells and type I IFN activated NK cells, contribute to dampening the efficacy of Ad vectors in individuals with pre-existing Ad immunity. In order to circumvent pre-existing immunity to adenovirus, numerous strategies, such as developing alternative Ad serotypes, varying immunization routes and utilizing prime-boost regimens, are under pre-clinical or clinical phases of development. However, these strategies mainly focus on one arm of pre-existing immunity. Selection of alternative serotypes has been largely driven by the absence in the human population of nAbs against them with little attention paid to cross-reactive Ad specific T cells. Conversely, varying the route of immunization appears to mainly rely on avoiding Ad specific tissue-resident T cells. Finally, prime-boost regimens do not actually circumvent pre-existing immunity but instead generate immune responses of sufficient magnitude to confer protection despite pre-existing immunity. Combining the above strategies and thus taking into account all components regulating pre-existing Ad immunity will help further improve the development of Ad vectors for animal and human use.
Collapse
|
6
|
Zhang P, Du E, Ma J, Wang W, Zhang L, Tikoo SK, Yang Z. A novel and simple method for rapid generation of recombinant porcine adenoviral vectors for transgene expression. PLoS One 2015; 10:e0127958. [PMID: 26011074 PMCID: PMC4444375 DOI: 10.1371/journal.pone.0127958] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 04/22/2015] [Indexed: 01/02/2023] Open
Abstract
Many human (different serotypes) and nonhuman adenovirus vectors are being used for gene delivery. However, the current system for isolating recombinant adenoviral vectors is either time-consuming or expensive, especially for the generation of recombinant non-human adenoviral vectors. We herein report a new and simple cloning approach for the rapid generation of a porcine adenovirus (PAdV-3) vector which shows promise for gene transfer to human cells and evasion of human adenovirus type 5 (HAdV-5) immunity. Based on the final cloning plasmid, pFPAV3-CcdB-Cm, and our modified SLiCE strategy (SLiCE cloning and lethal CcdB screening), the process for generating recombinant PAdV-3 plasmids required only one step in 3 days, with a cloning efficiency as high as 620 ± 49.56 clones/ng and zero background (100% accuracy). The recombinant PAdV-3 plasmids could be successfully rescued in porcine retinal pigment epithelium cells (VR1BL), which constitutively express the HAdV-5 E1 and PAdV-3 E1B 55k genes, and the foreign genes were highly expressed at 24 h after transduction into swine testicle (ST) cells. In conclusion, this strategy for generating recombinant PAdV-3 vectors based on our modified SLiCE cloning system was rapid and cost-efficient, which could be used as universal cloning method for modification the other regions of PAdV-3 genome as well as other adenoviral genomes.
Collapse
Affiliation(s)
- Peng Zhang
- College of Veterinary Medicine, North-west A&F University, Yangling, Shaanxi, China
| | - Enqi Du
- College of Veterinary Medicine, North-west A&F University, Yangling, Shaanxi, China
| | - Jing Ma
- College of Veterinary Medicine, North-west A&F University, Yangling, Shaanxi, China
| | - Wenbin Wang
- College of Veterinary Medicine, North-west A&F University, Yangling, Shaanxi, China
| | - Lu Zhang
- College of Veterinary Medicine, North-west A&F University, Yangling, Shaanxi, China
| | - Suresh K. Tikoo
- VIDO-InterVac, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- Vaccinology & Immunotherapeutics Program, School of Public Health, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- * E-mail: (ZY); (SKT)
| | - Zengqi Yang
- College of Veterinary Medicine, North-west A&F University, Yangling, Shaanxi, China
- * E-mail: (ZY); (SKT)
| |
Collapse
|
7
|
Lopez-Gordo E, Podgorski II, Downes N, Alemany R. Circumventing antivector immunity: potential use of nonhuman adenoviral vectors. Hum Gene Ther 2014; 25:285-300. [PMID: 24499174 DOI: 10.1089/hum.2013.228] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Adenoviruses are efficient gene delivery vectors based on their ability to transduce a wide variety of cell types and drive high-level transient transgene expression. While there have been advances in modifying human adenoviral (HAdV) vectors to increase their safety profile, there are still pitfalls that need to be further addressed. Preexisting humoral and cellular immunity against common HAdV serotypes limits the efficacy of gene transfer and duration of transgene expression. As an alternative, nonhuman AdV (NHAdV) vectors can circumvent neutralizing antibodies against HAdVs in immunized mice and monkeys and in human sera, suggesting that NHAdV vectors could circumvent preexisting humoral immunity against HAdVs in a clinical setting. Consequently, there has been an increased interest in developing NHAdV vectors for gene delivery in humans. In this review, we outline the recent advances and limitations of HAdV vectors for gene therapy and describe examples of NHAdV vectors focusing on their immunogenicity, tropism, and potential as effective gene therapy vehicles.
Collapse
Affiliation(s)
- Estrella Lopez-Gordo
- 1 Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow , Glasgow G12 8TA, United Kingdom
| | | | | | | |
Collapse
|
8
|
Duncan M, Cranfield MR, Torano H, Kuete HM, Lee GP, Glenn A, Bruder JT, Rangel D, Brough DE, Gall JG. Adenoviruses isolated from wild gorillas are closely related to human species C viruses. Virology 2013; 444:119-23. [PMID: 23806387 DOI: 10.1016/j.virol.2013.05.041] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Revised: 05/30/2013] [Accepted: 05/31/2013] [Indexed: 11/26/2022]
Abstract
We have isolated and cultured three distinct adenoviruses from wild gorillas. Phylogenetic analysis grouped the viruses with human adenovirus species C based on DNA polymerase, hexon, and E4ORF6 genes. The three wild gorilla adenoviruses clustered with the other species C captive gorilla adenoviruses, forming a branch separate from human and chimpanzee/bonobo adenoviruses. Animal sera to the three newly isolated viruses did not cross-neutralize, demonstrating serological distinctiveness. The human adenovirus 5 fiber knob blocked infection, suggesting use of the Coxsackie and Adenovirus Receptor. These viruses may provide viral vectors with properties distinct from chimpanzee adenovirus and human adenovirus vectors.
Collapse
Affiliation(s)
- McVey Duncan
- GenVec, Inc., 65W. Watkins Mill Rd, Gaithersburg, MD 20878, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
The development of gene-based vectors for immunization. Vaccines (Basel) 2013. [PMCID: PMC7151937 DOI: 10.1016/b978-1-4557-0090-5.00064-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
10
|
Abstract
Viral vectors have been developed as vaccine platforms for a number of pathogens and tumors. In particular, adenovirus (Ad)-based vectors expressing genes coding for pathogen or tumor antigens have proven efficacious to induce protective immunity. Major challenges in the use of Ad vectors are the high prevalence of anti-Ad immunity and the recent observation during an Ad-based HIV vaccine trial that led to increased HIV-1 acquisition in the presence of circulating anti-Ad5 neutralizing antibodies. In this review we summarize strategies to address these challenges and focus on modifications of the Ad capsid to enhance the adjuvant effect of anti-Ad immunogenicity and to circumvent pre-existing immunity. In addition, we summarize the current status and potential of other viral vector vaccines based on adeno-associated viruses, lentiviruses and poxviruses.
Collapse
|
11
|
Du E, Tikoo SK. Efficient replication and generation of recombinant bovine adenovirus-3 in nonbovine cotton rat lung cells expressing I-SceI endonuclease. J Gene Med 2011; 12:840-7. [PMID: 20963806 DOI: 10.1002/jgm.1505] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The rigorous evaluation of recombinant bovine adenovirus (BAdV)-3 as a gene delivery vector requires quick and efficient method of isolating recombinants. This requires both a suitable cell line and an efficient method of rescuing recombinant BAdV-3. To facilitate rapid isolation of recombinant BAdV-3, we have developed an efficient system for generating recombinants using newly identified nonbovine cell line permissive for replication of BAdV-3. METHODS Nonbovine cotton rat lung (CRL) cells in comparison to Madin-Darby bovine kidney cells and VIDO R2 cells were analyzed for the production of progeny virus and DNA transfection efficiency. In addition, lentiviral expression system was used to generate stable nonbovine CRL cell line expressing endonuclease I-SceI as examined by western blotting. Transfection of this cell line with circular or linear plasmid containing full-length BAdV-3 genome was used to generate recombinant BAdV-3. RESULTS We demonstrate that nonbovine CRL cells are permissive for replication of BAdV-3 and can be efficiently transfected with plasmid DNA. Second, we constructed CRL cell line (VIDO DT1) expressing an intron-encoding endonuclease I-SceI. Finally, we demonstrate that transfection of VIDO DT1 cells with a circular plasmid containing recombinant BAdV-3 genome flanked by I-SceI recognition sites can efficiently rescue recombinant virus. CONCLUSIONS The use of circular molecular clones together with I-SceI endonuclease expressing, BAdV-3 permissive CRL cell line not only increased the viral genome transfection efficiency, but also reduced the viral rescue time and amount of DNA required for rescuing recombinant BAdV-3s.
Collapse
Affiliation(s)
- Enqi Du
- Vaccine and Infectious Disease Organization, University of Saskatchewan Saskatoon, Saskatchewan, Canada
| | | |
Collapse
|
12
|
Krause A, Whu WZ, Xu Y, Joh J, Crystal RG, Worgall S. Protective anti-Pseudomonas aeruginosa humoral and cellular mucosal immunity by AdC7-mediated expression of the P. aeruginosa protein OprF. Vaccine 2011; 29:2131-9. [PMID: 21215829 DOI: 10.1016/j.vaccine.2010.12.087] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2010] [Revised: 12/09/2010] [Accepted: 12/20/2010] [Indexed: 10/18/2022]
Abstract
Replication-deficient adenoviral (Ad) vectors are an attractive platform for a vaccine against lung infections caused by Pseudomonas aeruginosa. Ad vectors based on non-human serotypes have been developed to circumvent the problem of pre-existing anti-Ad immunity in humans. The present study analyzes the anti-P. aeruginosa systemic and lung mucosal immunity elicited by a non-human primate-based AdC7 vector expressing the outer membrane protein F (AdC7OprF) of P. aeruginosa. Intramuscular immunization of mice with AdC7OprF induced similar levels of serum and mucosal anti-OprF IgG and increased levels of anti-OprF IgA in lung epithelial lining fluid (ELF) compared to immunization with a human serotype Ad5OprF vector (p>0.05). OprF-specific INF-γ in splenic T cells stimulated with OprF-pulsed syngeneic splenic dendritic cells (DC) was similar following immunization with AdC7OprF compared to Ad5OprF (p>0.05). In contrast, OprF-specific INF-γ responses in lung T cells stimulated with either spleen or lung DC were increased following immunization with AdC7OprF compared to Ad5OprF (p<0.05). Interestingly, direct administration of AdC7OprF to the respiratory tract resulted in an increase of OprF-specific IgG in serum, OprF-specific IgG and IgA in lung ELF, and OprF-specific INF-γ in lung T-cells compared to immunization with Ad5OprF, and survival following challenge with a lethal dose of P. aeruginosa. These data demonstrate that systemic or lung mucosal immunization with an AdC7-based vaccine vector induces superior pulmonary humoral and cellular anti-transgene immunity compared to immunization with an Ad5-based vector and favors AdC7-based vectors as vaccines to induce lung mucosal immunity.
Collapse
Affiliation(s)
- Anja Krause
- Department of Genetic Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | | | | | | | | | | |
Collapse
|
13
|
Bru T, Salinas S, Kremer EJ. An update on canine adenovirus type 2 and its vectors. Viruses 2010; 2:2134-2153. [PMID: 21994722 PMCID: PMC3185752 DOI: 10.3390/v2092134] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2010] [Revised: 09/23/2010] [Accepted: 09/24/2010] [Indexed: 12/27/2022] Open
Abstract
Adenovirus vectors have significant potential for long- or short-term gene transfer. Preclinical and clinical studies using human derived adenoviruses (HAd) have demonstrated the feasibility of flexible hybrid vector designs, robust expression and induction of protective immunity. However, clinical use of HAd vectors can, under some conditions, be limited by pre-existing vector immunity. Pre-existing humoral and cellular anti-capsid immunity limits the efficacy and duration of transgene expression and is poorly circumvented by injections of larger doses and immuno-suppressing drugs. This review updates canine adenovirus serotype 2 (CAV-2, also known as CAdV-2) biology and gives an overview of the generation of early region 1 (E1)-deleted to helper-dependent (HD) CAV-2 vectors. We also summarize the essential characteristics concerning their interaction with the anti-HAd memory immune responses in humans, the preferential transduction of neurons, and its high level of retrograde axonal transport in the central and peripheral nervous system. CAV-2 vectors are particularly interesting tools to study the pathophysiology and potential treatment of neurodegenerative diseases, as anti-tumoral and anti-viral vaccines, tracer of synaptic junctions, oncolytic virus and as a platform to generate chimeric vectors.
Collapse
Affiliation(s)
- Thierry Bru
- Institut de Génétique Moléculaire de Montpellier, CNRS UMR 5535, 1919 Route de Mende Montpellier, 34293 France; E-Mails: (T.B.); (S.S.)
- Université de Montpellier I, 5 Bd Henri IV, 34000 Montpellier, France
- Université de Montpellier II, place Eugène Bataillon, 34090 Montpellier, France
| | - Sara Salinas
- Institut de Génétique Moléculaire de Montpellier, CNRS UMR 5535, 1919 Route de Mende Montpellier, 34293 France; E-Mails: (T.B.); (S.S.)
- Université de Montpellier I, 5 Bd Henri IV, 34000 Montpellier, France
- Université de Montpellier II, place Eugène Bataillon, 34090 Montpellier, France
| | - Eric J. Kremer
- Institut de Génétique Moléculaire de Montpellier, CNRS UMR 5535, 1919 Route de Mende Montpellier, 34293 France; E-Mails: (T.B.); (S.S.)
- Université de Montpellier I, 5 Bd Henri IV, 34000 Montpellier, France
- Université de Montpellier II, place Eugène Bataillon, 34090 Montpellier, France
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +33-467-613-372; Fax: +33-467-040-231
| |
Collapse
|
14
|
Induction of both cellular and humoral immunity following a rational prime-boost immunization regimen that incorporates recombinant ovine atadenovirus and fowlpox virus. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2010; 17:1679-86. [PMID: 20810681 DOI: 10.1128/cvi.00291-10] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Recombinant fowlpox viruses (rFPV) and ovine atadenoviruses (rOAdV) are being developed as safe, nonpathogenic, prophylactic and therapeutic vaccine vectors. There is scope, however, to improve the limited immune responses elicited by each of these vaccine vectors. Using previously determined and optimized routes of administration and viral doses, we characterized the primary adaptive immune responses elicited by recombinant variants of each virus. We demonstrate the contrasting nature of the response elicited by each recombinant virus. Whereas rFPV generates predominately cell-mediated immunity to our nominal target antigen, ovalbumin (OVA), rOAdV drives strong humoral responses. By defining the time taken to achieve maximal cytotoxic T cell responses and by studying the different patterns and kinetics of major histocompatibility complex class I-restricted OVA antigen expression postimmunization, we proposed a heterologous prime-boost regimen of immunization with rOAdV followed by rFPV. The subsequent experimental results showed that this approach produced robust cell-mediated and humoral immune responses against OVA that, importantly, were accompanied by weak anti-viral vector antibody responses. These results, therefore, represent a novel and potentially clinically applicable way to achieve broadly based and effective immunity to the antigens encoded by vectored vaccines.
Collapse
|
15
|
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: 1.3] [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
|
16
|
Zaiss AK, Machado HB, Herschman HR. The influence of innate and pre-existing immunity on adenovirus therapy. J Cell Biochem 2010; 108:778-90. [PMID: 19711370 DOI: 10.1002/jcb.22328] [Citation(s) in RCA: 111] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Recombinant adenovirus serotype 5 (Ad5) vectors have been studied extensively in preclinical gene therapy models and in a range of clinical trials. However, innate immune responses to adenovirus vectors limit effectiveness of Ad5 based therapies. Moreover, extensive pre-existing Ad5 immunity in human populations will likely limit the clinical utility of adenovirus vectors, unless methods to circumvent neutralizing antibodies that bind virus and block target cell transduction can be developed. Furthermore, memory T cell and humoral responses to Ad5 are associated with increased toxicity, raising safety concerns for therapeutic adenovirus vectors in immunized hosts. Most preclinical studies have been performed in naïve animals; although pre-existing immunity is among the greatest hurdles for adenovirus therapies, it is also one of the most neglected experimentally. Here we summarize findings using adenovirus vectors in naïve animals, in Ad-immunized animals and in clinical trials, and review strategies proposed to overcome innate immune responses and pre-existing immunity.
Collapse
Affiliation(s)
- Anne K Zaiss
- Department of Biological Chemistry, David Geffen School of Medicine, UCLA, Los Angeles, California 90095, USA
| | | | | |
Collapse
|
17
|
Corredor JC, Nagy E. The non-essential left end region of the fowl adenovirus 9 genome is suitable for foreign gene insertion/replacement. Virus Res 2010; 149:167-74. [PMID: 20132849 DOI: 10.1016/j.virusres.2010.01.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2009] [Revised: 01/26/2010] [Accepted: 01/26/2010] [Indexed: 11/26/2022]
Abstract
The goals of this study were to demonstrate that a non-essential region at the left end of the fowl adenovirus 9 (FAdV-9) genome could be used to generate recombinant viruses, examine their in vitro growth characteristics and determine their ability to transduce non-avian cells. Three FAdV-9 vectors (rFAdV-9s) were generated carrying the enhanced-green fluorescent protein (EGFP) gene: FAdV-9inEGFP, FAdV-9 Delta 1-EGFP and FAdV-9 Delta 4-EGFP. FAdV-9inEGFP carried the EGFP cassette inserted into the non-essential region without deletion resulting in an increase of the genome size to 103.7% of the wild-type. FAdV-9 Delta 1-EGFP and FAdV-9 Delta 4-EGFP (rFAdV-9 Delta s) carried the EGFP cassette replacing the non-essential sequences at nucleotides 1194-2342 and 491-2782, respectively. All rFAdV-9s had wild-type growth kinetics and plaque morphology. The rFAdV-9 Delta s replicated in CH-SAH cells with the same titers as the wild-type virus. The FAdV-9inEGFP titers were approximately 1 log lower than those of rFAdV-9 Delta s and wt FAdV-9 at 36 and 48 h post-infection (h.p.i.). EGFP was expressed in avian and mammalian cells infected with rFAdV-9s. EGFP expression, based on spectrofluorometry, was significantly higher in chicken hepatoma cells infected with FAdV-9inEGFP than in those with rFAdV-9 Delta s at 18 and 24h.p.i, suggesting a functional role of some or all non-essential ORFs on foreign gene expression. This study demonstrated the suitability of the non-essential region as an insertion/replacement site for foreign genes to generate FAdV-9-based vectors that can be applied as recombinant vaccines for poultry or gene delivery vehicles for mammalian systems.
Collapse
Affiliation(s)
- Juan Carlos Corredor
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, 50 Stone Road East, Guelph, Ontario, N1G 2W1 Canada
| | | |
Collapse
|
18
|
Bridgeman A, Roshorm Y, Lockett LJ, Xu ZZ, Hopkins R, Shaw J, Both GW, Hanke T. Ovine atadenovirus, a novel and highly immunogenic vector in prime-boost studies of a candidate HIV-1 vaccine. Vaccine 2009; 28:474-83. [PMID: 19853074 DOI: 10.1016/j.vaccine.2009.09.136] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2009] [Revised: 09/28/2009] [Accepted: 09/30/2009] [Indexed: 02/08/2023]
Abstract
Ovine adenovirus type 7 (OAdV) is the prototype member of the genus Atadenovirus. No immunity to the virus has so far been detected in human sera. We describe the construction and evaluation of a candidate HIV-1 vaccine based on OAdV and its utilisation alone and in combination with plasmid-, human adenovirus type 5 (HAdV5; a Mastadenovirus)-, and modified vaccinia Ankara (MVA)-vectored vaccines. All vectors expressed HIVA, an immunogen consisting of HIV-1 clade A consensus Gag-derived protein coupled to a T cell polyepitope. OAdV.HIVA was genetically stable, grew well and expressed high levels of protein from the Rous sarcoma virus promoter. OAdV.HIVA was highly immunogenic in mice and efficiently primed and boosted HIV-1-specific T cell responses together with heterologous HIVA-expressing vectors. There were significant differences between OAdV and HAdV5 vectors in priming of naïve CD8(+) T cell responses to HIVA and in the persistence of MHC class I-restricted epitope presentation in the local draining lymph nodes. OAdV.HIVA primed T cells more rapidly but was less persistent than AdV5.HIVA and thus induced a qualitatively distinct T cell response. Nevertheless, both vectors primed a response in mice that reduced viral titres in a surrogate challenge model by three to four orders of magnitude. Thus, OAdV is a novel, underexplored vaccine vector with potential for further development for HIV-1 and other vaccines. The data are discussed in the context of the latest HIV-1 vaccine developments.
Collapse
Affiliation(s)
- Anne Bridgeman
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe, Oxford OX3 9DS, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Abstract
The antiquated system used to manufacture the currently licensed inactivated influenza virus vaccines would not be adequate during an influenza virus pandemic. There is currently a search for vaccines that can be developed faster and provide superior, long-lasting immunity to influenza virus as well as other highly pathogenic viruses and bacteria. Recombinant vectors provide a safe and effective method to elicit a strong immune response to a foreign protein or epitope. This review explores the advantages and limitations of several different vectors that are currently being tested, and highlights some of the newer viruses being used as recombinant vectors.
Collapse
|
20
|
Comparative analysis of vector biodistribution, persistence and gene expression following intravenous delivery of bovine, porcine and human adenoviral vectors in a mouse model. Virology 2009; 386:44-54. [PMID: 19211122 DOI: 10.1016/j.virol.2009.01.008] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2008] [Revised: 11/03/2008] [Accepted: 01/09/2009] [Indexed: 11/23/2022]
Abstract
Nonhuman adenoviruses including bovine adenovirus serotype 3 (BAd3) and porcine adenovirus serotype 3 (PAd3) can circumvent pre-existing immunity against human adenovirus serotype 5 (HAd5) and are being developed as alternative vectors for gene delivery. To assess the usefulness of these vectors for in vivo gene delivery, we compared biodistribution, persistence, state of vector genome, and transgene and vector gene expression by replication-defective BAd3 and PAd3 vectors with those of HAd5 vector in a FVB/n mouse model following intravenous inoculation. BAd3 vector efficiently transduced the heart, kidney and lung in addition to the liver and spleen and persisted for a longer duration compared to PAd3 or HAd5 vectors. Biodistribution of PAd3 vector was comparable to that of HAd5 vector but showed more rapid vector clearance. Only linear episomal forms of BAd3, PAd3, and HAd5 vector genomes were detected. All three vectors efficiently expressed the green fluorescent protein (GFP) transgene proportionate to the vector genome copy number in various tissues. Furthermore, leaky expression of vector genes, both the early (E4) and the late (hexon) was observed in all three vectors and gradually declined with time. These results suggest that BAd3 and PAd3 vectors could serve as an alternative or supplement to HAd5 for gene delivery applications.
Collapse
|
21
|
Schoenly KA, Weiner DB. Human immunodeficiency virus type 1 vaccine development: recent advances in the cytotoxic T-lymphocyte platform "spotty business". J Virol 2008; 82:3166-80. [PMID: 17989174 PMCID: PMC2268479 DOI: 10.1128/jvi.01634-07] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Kimberly A Schoenly
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | | |
Collapse
|
22
|
Singh N, Pandey A, Jayashankar L, Mittal SK. Bovine adenoviral vector-based H5N1 influenza vaccine overcomes exceptionally high levels of pre-existing immunity against human adenovirus. Mol Ther 2008; 16:965-71. [PMID: 18301400 DOI: 10.1038/mt.2008.12] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Because of the high prevalence of adenovirus (Ad) infections in humans, it is believed that pre-existing Ad-neutralizing antibodies (vector immunity) may negatively impact the immune response to vaccine antigens when delivered by human Ad (HAd) vectors. In order to evaluate whether bovine Ad subtype 3 (BAd3), a non-HAd vector, can effectively elude high levels of pre-existing vector immunity, naïve and HAd serotype 5 (HAd)-primed mice were immunized with BAd-H5HA [BAd3 vector expressing the hemagglutinin (HA) gene from H5N1 influenza virus]. Even in the presence of very high levels of HAd-specific neutralizing antibody, no significant reductions in HA-specific humoral and cell-mediated immune (CMI) responses were observed in HAd-primed mice immunized with BAd-H5HA. In naïve mice immunized with HAd-H5HA (HAd5 vector expressing H5N1 HA) and boosted with BAd-H5HA, the humoral responses elicited were significantly higher (P < 0.01) than with either HAd-H5HA or BAd-H5HA alone, while the CMI responses were comparable in the groups. This finding underlines the importance of a heterologous prime-boost approach for achieving an enhanced immune response. The immunization of naïve or HAd-primed mice with BAd-H5HA bestowed full protection from morbidity and mortality following a potentially lethal challenge with A/Hong Kong/483/97. These results demonstrate the importance of BAd vectors as an alternate or supplement to HAd vectors for influenza pandemic preparedness.
Collapse
Affiliation(s)
- Neetu Singh
- Department of Comparative Pathobiology, Purdue University, West Lafayette, Indiana 47907, USA
| | | | | | | |
Collapse
|
23
|
Nabel GJ. The development of gene-based vectors for immunization. Vaccines (Basel) 2008. [PMCID: PMC7310921 DOI: 10.1016/b978-1-4160-3611-1.50066-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
|
24
|
Yang TC, Millar JB, Grinshtein N, Bassett J, Finn J, Bramson JL. T-cell immunity generated by recombinant adenovirus vaccines. Expert Rev Vaccines 2007; 6:347-56. [PMID: 17542750 DOI: 10.1586/14760584.6.3.347] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Recombinant adenovirus vaccines show great promise for generating protective immunity against infectious agents and tumors. Our studies have identified several interesting biological features of the adenovirus vector that influence the T-cell response. Notably, we have demonstrated that following immunization with adenovirus vaccines, the transgene antigen remains available to the system for a longer period than would be expected, resulting in a T-cell population with a sustained effector phenotype. The implications of these observations with regards to the utility of adenovirus vaccines are discussed.
Collapse
Affiliation(s)
- Teng Chih Yang
- Center for Gene Therapeutics, Department of Pathology and Molecular Medicine, McMaster University, 1200 Main Street West, Hamilton, Ontario, L8N 3Z5, Canada.
| | | | | | | | | | | |
Collapse
|
25
|
Perreau M, Guérin MC, Drouet C, Kremer EJ. Interactions between human plasma components and a xenogenic adenovirus vector: reduced immunogenicity during gene transfer. Mol Ther 2007; 15:1998-2007. [PMID: 17712332 DOI: 10.1038/sj.mt.6300289] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
By the time we are adolescents most of us have been in contact with several of the >50 human adenovirus (HAd) serotypes. These common subclinical infections lead to an efficient anti-adenovirus cross-reacting adaptive immunity. During gene therapy, the ubiquitous anti-adenovirus humoral response and complement activation will modify and dictate vector biodistribution, as well as the response to the virion and transgene(s). In this study, we assayed the interactions of a xenogenic adenovirus derived from canine serotype 2 (CAV-2) with naturally occurring human antibodies (Abs) and the complement system. In our cohort, we found class G immunoglobulins (Igs) that recognized the intact CAV-2 virion and the external virion proteins. However, the majority of donors had low or no neutralizing Abs, class A, or class M Igs. Purified anti-HAd serotype 5 Abs also recognized CAV-2 virion proteins. In addition, in spite of the presence of anti-CAV-2 IgGs, CAV-2 poorly activated the classical and alternative complement cascades. This atypical response was due to a block upstream of the component 3 (C3) convertase and interplay between the component 1 (C1) inhibitor, the C1q-C1r2-C1s2 complex and CAV-2. Our data demonstrate that some xenogenic adenovirus vectors, like CAV-2, could lead to notably different outcomes following systemic delivery.
Collapse
Affiliation(s)
- Matthieu Perreau
- Institut de Génétique Moléculaire de Montpellier, Montpellier, France
| | | | | | | |
Collapse
|
26
|
He Y, Falo LD. Induction of T cell immunity by cutaneous genetic immunization with recombinant lentivector. Immunol Res 2007; 36:101-17. [PMID: 17337771 PMCID: PMC3065303 DOI: 10.1385/ir:36:1:101] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/1999] [Revised: 11/30/1999] [Accepted: 11/30/1999] [Indexed: 01/12/2023]
Abstract
Recent studies suggest that T cell-based cellular immunity plays an important role in preventing and delaying progression of infectious and neoplastic diseases. Based on these findings, recent efforts in vaccine research are giving rise to a new generation of "T cell" vaccines. The development of T cell vaccines has been problematic. Current investigations are focusing on gene-based immunization strategies, including the development of non-viral "naked" plasmid DNA and recombinant viral vector-based genetic immunization approaches. Here, we briefly review recent progress in the development of recombinant viral vectors for genetic immunization and our own recent studies elucidating differences in mechanisms of genetic immunization. We propose that the mechanism of immune induction depends in part on unique features of specific viral vectors, and that a comparison of representative vectors mechanistically will enable a more informed understanding of the determining parameters of immune induction. Our initial studies have focused on the identification of antigen-presenting-cell subsets important for priming CD8+ T cell immunity, the effects of antigen persistence on immune responses, and the unique immunogenicity of skin as a target tissue for vaccine delivery. We review data suggesting that the unique properties of recombinant lentivectors make them appealing candidates as genetic immunization vehicles for eliciting T cell immune responses.
Collapse
Affiliation(s)
- Yukai He
- Department of Dermatology, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA.
| | | |
Collapse
|
27
|
Abbink P, Lemckert AAC, Ewald BA, Lynch DM, Denholtz M, Smits S, Holterman L, Damen I, Vogels R, Thorner AR, O'Brien KL, Carville A, Mansfield KG, Goudsmit J, Havenga MJE, Barouch DH. Comparative seroprevalence and immunogenicity of six rare serotype recombinant adenovirus vaccine vectors from subgroups B and D. J Virol 2007; 81:4654-63. [PMID: 17329340 PMCID: PMC1900173 DOI: 10.1128/jvi.02696-06] [Citation(s) in RCA: 385] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Recombinant adenovirus serotype 5 (rAd5) vector-based vaccines are currently being developed for both human immunodeficiency virus type 1 and other pathogens. The potential limitations associated with rAd5 vectors, however, have led to the construction of novel rAd vectors derived from rare Ad serotypes. Several rare serotype rAd vectors have already been described, but a detailed comparison of multiple rAd vectors from subgroups B and D has not previously been reported. Such a comparison is critical for selecting optimal rAd vectors for advancement into clinical trials. Here we describe the construction of three novel rAd vector systems from Ad26, Ad48, and Ad50. We report comparative seroprevalence and immunogenicity studies involving rAd11, rAd35, and rAd50 vectors from subgroup B; rAd26, rAd48, and rAd49 vectors from subgroup D; and rAd5 vectors from subgroup C. All six rAd vectors from subgroups B and D exhibited low seroprevalence in a cohort of 200 individuals from sub-Saharan Africa, and they elicited Gag-specific cellular immune responses in mice both with and without preexisting anti-Ad5 immunity. The rAd vectors from subgroup D were also evaluated using rhesus monkeys and were shown to be immunogenic after a single injection. The rAd26 vectors proved the most immunogenic among the rare serotype rAd vectors studied, although all rare serotype rAd vectors were still less potent than rAd5 vectors in the absence of anti-Ad5 immunity. These studies substantially expand the portfolio of rare serotype rAd vectors that may prove useful as vaccine vectors for the developing world.
Collapse
Affiliation(s)
- Peter Abbink
- Research East Room 213, Division of Viral Pathogenesis, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Stevenson M, Boos E, Herbert C, Hale A, Green N, Lyons M, Chandler L, Ulbrich K, van Rooijen N, Mautner V, Fisher K, Seymour L. Chick embryo lethal orphan virus can be polymer-coated and retargeted to infect mammalian cells. Gene Ther 2006; 13:356-68. [PMID: 16355117 DOI: 10.1038/sj.gt.3302655] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Non-human adenovirus vectors have attractive immunological properties for gene therapy but are frequently restricted by inefficient transduction of human target cells. Using chicken embryo lethal orphan (CELO) virus, we employed a nongenetic mechanism of polymer coating and retargeting with basic fibroblast growth factor (bFGF-pc-CELOluc), a strategy that permits efficient tropism modification of human adenovirus. bFGF-pc-CELOluc showed efficient uptake and transgene expression in chick embryo fibroblasts (CEF), and increased levels of binding and internalization in a variety of human cell lines. Transgene expression was also greater than unmodified CELOluc in PC-3 human prostate cells, although the specific activity (RLU per internalized viral genome) was decreased. In CEF, the specific activity of bFGF-pc-CELOluc was considerably higher than in the human prostate cell line PC-3. Retargeted virus was fully resistant to inhibition by human serum with known adenovirus-neutralizing activity in vitro, while in mice CELOluc was cleared less rapidly from the blood than Adluc following i.v. administration in the presence of adenovirus neutralizing serum. Polymer coating and retargeting with bFGF further reduced rates of clearance for both viruses, suggesting protection against both neutralizing and opsonizing factors. The data indicate that CELO virus may be retargeted to infect human cells via alternative, potentially disease-specific, receptors and resist the effects of pre-existing humoral immunity.
Collapse
Affiliation(s)
- M Stevenson
- Department of Clinical Pharmacology, University of Oxford, Oxford, UK.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Sakurai F, Kawabata K, Koizumi N, Inoue N, Okabe M, Yamaguchi T, Hayakawa T, Mizuguchi H. Adenovirus serotype 35 vector-mediated transduction into human CD46-transgenic mice. Gene Ther 2006; 13:1118-26. [PMID: 16541121 DOI: 10.1038/sj.gt.3302749] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
We previously demonstrated that systemic administration of adenovirus serotype 35 (Ad35) vectors to mice does not mediate efficient transduction in organs, probably because expression of the mouse analog of the subgroup B Ad receptor, human CD46 (membrane cofactor protein), is limited to the testis. Here, we describe the in vitro and in vivo transduction characteristics of Ad35 vectors by using homozygous and hemizygous human CD46-transgenic (CD46TG) mice, which ubiquitously express human CD46. An Ad35 vector more efficiently transduced the primary dendritic cells and macrophages prepared from CD46TG mice than those from wild-type mice. In vivo transduction experiments demonstrated that CD46TG mice are more susceptible to Ad35 vector-mediated in vivo transduction than are wild-type mice. In particular, homozygous CD46TG mice, which express higher levels of CD46 in the organs than hemizygous CD46TG mice, tend to exhibit higher transduction efficiencies after intraperitoneal administration than hemizygous CD46TG mice. Intraperitoneal administration of Ad35 vectors resulted in efficient transduction into the mesothelial cells of the peritoneal organs in homozygous CD46TG mice. These results indicate that an Ad35 vector recognizes human CD46 as a cellular receptor in CD46TG mice. However, the in vivo transduction efficiencies of Ad35 vectors in CD46TG mice are much lower than those of conventional Ad5 vectors in wild-type mice.
Collapse
Affiliation(s)
- F Sakurai
- Laboratory of Gene Transfer and Regulation, National Institute of Biomedical Innovation, Osaka, Japan
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Xin KQ, Jounai N, Someya K, Honma K, Mizuguchi H, Naganawa S, Kitamura K, Hayakawa T, Saha S, Takeshita F, Okuda K, Honda M, Klinman DM, Okuda K. Prime-boost vaccination with plasmid DNA and a chimeric adenovirus type 5 vector with type 35 fiber induces protective immunity against HIV. Gene Ther 2006; 12:1769-77. [PMID: 16079886 DOI: 10.1038/sj.gt.3302590] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Immunization involving a DNA vaccine prime followed by an adenovirus type 5 (Ad5) boost elicited a protective immune response against SHIV challenge in monkeys. However, the hepatocellular tropism of Ad5 limits the safety of this viral vector. This study examines the safety and immunogenicity of a replication-defective chimeric Ad5 vector with the Ad35 fiber (Ad5/35) in BALB/c mice and rhesus monkeys. This novel Ad5/35 vector showed minimal hepatotoxicity after intramuscular administration with the novel Ad5/35 vector. In addition, an Ad5/35 vector expressing HIV Env gp160 protein (Ad5/35-HIV) generated strong HIV-specific immune responses in both animal models. Priming with a DNA vaccine followed by Ad5/35-HIV boosting yielded protection against a gp160-expressing vaccinia virus challenge in BALB/c mice. The Ad5/35-HIV vector was significantly less susceptible to the pre-existing Ad5 immunity than a comparable Ad5 vector. These findings indicate that an Ad5/35 vector-based HIV vaccine may be of considerable value for clinical use.
Collapse
Affiliation(s)
- K-Q Xin
- Department of Molecular Biodefense Research, Yokohama City University, Graduate School of Medicine, Yokohama, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
Vaccine development has traditionally been an empirical discipline. Classical vaccine strategies include the development of attenuated organisms, whole killed organisms, and protein subunits, followed by empirical optimization and iterative improvements. While these strategies have been remarkably successful for a wide variety of viruses and bacteria, these approaches have proven more limited for pathogens that require cellular immune responses for their control. In this review, current strategies to develop and optimize gene-based vaccines are described, with an emphasis on novel approaches to improve plasmid DNA vaccines and recombinant adenovirus vector-based vaccines.
Collapse
Affiliation(s)
- Dan H Barouch
- Research East Room 113, Division of Viral Pathogenesis, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA.
| |
Collapse
|
32
|
Nakayama M, Both GW, Banizs B, Tsuruta Y, Yamamoto S, Kawakami Y, Douglas JT, Tani K, Curiel DT, Glasgow JN. An adenovirus serotype 5 vector with fibers derived from ovine atadenovirus demonstrates CAR-independent tropism and unique biodistribution in mice. Virology 2006; 350:103-15. [PMID: 16516257 DOI: 10.1016/j.virol.2006.01.037] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2005] [Revised: 12/23/2005] [Accepted: 01/26/2006] [Indexed: 01/09/2023]
Abstract
Many clinically important tissues are refractory to adenovirus (Ad) infection due to negligible levels of the primary Ad5 receptor the coxsackie and adenovirus receptor CAR. Thus, development of novel CAR-independent Ad vectors should lead to therapeutic gain. Ovine atadenovirus type 7, the prototype member of genus Atadenovirus, efficiently transduces CAR-deficient human cells in vitro, and systemic administration of OAdV is not associated with liver sequestration in mice. The penton base of OAdV7 does not contain an RGD motif, implicating the long-shafted fiber molecule as a major structural dictate of OAdV tropism. We hypothesized that replacement of the Ad5 fiber with the OAdV7 fiber would result in an Ad5 vector with CAR-independent tropism in vitro and liver "detargeting" in vivo. An Ad5 vector displaying the OAdV7 fiber was constructed (Ad5Luc1-OvF) and displayed CAR-independent, enhanced transduction of CAR-deficient human cells. When administered systemically to C57BL/6 mice, Ad5Luc1-OvF reporter gene expression was reduced by 80% in the liver compared to Ad5 and exhibited 50-fold higher gene expression in the kidney than the control vector. To our knowledge, this is the first report of a fiber-pseudotyped Ad vector that simultaneously displays decreased liver uptake and a distinct organ tropism in vivo. This vector may have future utility in murine models of renal disease.
Collapse
Affiliation(s)
- Masaharu Nakayama
- Division of Human Gene Therapy, Department of Medicine, University of Alabama at Birmingham, 901 19th Street South BMR2-572, Birmingham, AL 35294-2180, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Gallo P, Dharmapuri S, Cipriani B, Monaci P. Adenovirus as vehicle for anticancer genetic immunotherapy. Gene Ther 2006; 12 Suppl 1:S84-91. [PMID: 16231059 DOI: 10.1038/sj.gt.3302619] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Adenoviruses (Ads) are in the forefront of genetic immunization methods being developed against cancer. Their ability to elicit an effective immune response against tumor-associated antigens has been demonstrated in many model systems. Several clinical trials, which use Ad as vehicle for immunization, are already in progress. Preclinical studies have also demonstrated the efficacy of combining Ad-mediated immunization with adjuvants such as chemotherapeutic agents and cytokines. Issues related to sero-prevalence and safety of Ads, however, continue to pose a challenge and need to be addressed.
Collapse
Affiliation(s)
- P Gallo
- Department of Molecular and Cell Biology, I.R.B.M.P. Angeletti, Pomezia, Roma, Italy
| | | | | | | |
Collapse
|
34
|
Boyer JL, Crystal RG. Genetic medicine strategies to protect against bioterrorism. TRANSACTIONS OF THE AMERICAN CLINICAL AND CLIMATOLOGICAL ASSOCIATION 2006; 117:297-310; discussion 310-1. [PMID: 18528482 PMCID: PMC1500914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Affiliation(s)
- Julie L Boyer
- Department of Genetic Medicine, Weill Medical College of Cornell University, New York, New York 10021, USA
| | | |
Collapse
|
35
|
Appaiahgari MB, Saini M, Rauthan M, Vrati S. Immunization with recombinant adenovirus synthesizing the secretory form of Japanese encephalitis virus envelope protein protects adenovirus-exposed mice against lethal encephalitis. Microbes Infect 2006; 8:92-104. [PMID: 16126425 DOI: 10.1016/j.micinf.2005.05.023] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2005] [Accepted: 05/31/2005] [Indexed: 11/21/2022]
Abstract
Replication-defective recombinant adenoviruses (RAds) were constructed that synthesized the pre-membrane and envelope (E) proteins of Japanese encephalitis virus (JEV). Recombinant virus RAdEa synthesized Ea, the membrane-anchored E protein, and RAdEs synthesized Es, the secretory E protein. Compared with RAdEs, RAdEa replicated poorly in HEK 293A cells and synthesized lower amounts of E protein. Oral immunization of mice with RAds generated low titers of anti-JEV antibodies that had little JEV neutralizing activity. Intra-muscular (IM) immunization of mice with either RAd generated high titers of anti-JEV antibodies. Interestingly, RAdEa induced only low titers of JEV neutralizing antibodies. Titers were significantly higher in case of RAdEs immunization. Splenocytes from mice immunized IM with RAds secreted large amounts of interferon-gamma and moderate amounts of interleukin-5 in the presence of JEV and showed cytotoxic activity against JEV-infected cells. Naïve mice immunized IM with RAdEs showed complete protection against a lethal dose of JEV given intra-cerebrally. In order to study the effect of the pre-existing adenovirus 5 (Ad5) immunity on the outcome of the RAdEs immunization, mice were exposed to Ad5 through IM or intra-nasal (IN) routes before immunization with RAdEs. Mice exposed to Ad5 through the IN route, when immunized with RAdEs given IM, or those exposed to Ad5 through the IM route, when immunized with RAdEs given IN, were completely protected against lethal JEV challenge.
Collapse
Affiliation(s)
- Mohan Babu Appaiahgari
- Virology Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, JNU Complex, New Delhi 110 067, India
| | | | | | | |
Collapse
|
36
|
Hashimoto M, Boyer JL, Hackett NR, Wilson JM, Crystal RG. Induction of protective immunity to anthrax lethal toxin with a nonhuman primate adenovirus-based vaccine in the presence of preexisting anti-human adenovirus immunity. Infect Immun 2005; 73:6885-91. [PMID: 16177368 PMCID: PMC1230977 DOI: 10.1128/iai.73.10.6885-6891.2005] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Prevention or therapy for bioterrorism-associated anthrax infections requires rapidly acting effective vaccines. We recently demonstrated (Y. Tan, N. R. Hackett, J. L. Boyer, and R. G. Crystal, Hum. Gene Ther. 14:1673-1682, 2003) that a single administration of a recombinant serotype 5 adenovirus (Ad) vector expressing anthrax protective antigen (PA) provides rapid protection against anthrax lethal toxin challenge. However, approximately 35 to 50% of humans have preexisting neutralizing antibodies against Ad5. This study assesses the hypothesis that a recombinant adenovirus vaccine based on the nonhuman primate-derived serotype AdC7, against which humans do not have immunity, expressing PA (AdC7PA) will protect against anthrax lethal toxin even in the presence of preexisting anti-Ad5 immunity. Naive and Ad5-immunized BALB/c mice received (intramuscularly) 10(8) to 10(11) particle units (PU) of AdC7PA, Ad5PA (a human serotype Ad5-based vector expressing a secreted form of PA), or AdNull (an Ad5 vector with no transgene). Robust anti-PA immunoglobulin G and neutralizing antibodies were detected by 2 to 4 weeks following administration of AdC7PA to naive or Ad5 preimmunized mice, whereas low anti-PA titers were detected in Ad5-preimmunized mice following administration of Ad5PA. To assess protection in vivo, naive or mice previously immunized against Ad5 were immunized with AdC7PA or Ad5PA and then challenged with a lethal intravenous dose of Bacillus anthracis lethal toxin. Whereas Ad5PA protected naive mice against challenge with B. anthracis lethal toxin, Ad5PA was ineffective in mice that were previously immunized against Ad5. In contrast, AdC7PA functioned effectively not only to protect naive mice but also to protect Ad5-preimmunized mice, with 100% survival after lethal toxin challenge. These data suggest the nonhuman-based vector AdC7PA is an effective vaccine for the development of protective immunity against B. anthracis and importantly functions as a "sero-switch" base for an adenovirus vaccine to function in the context of preexisting anti-Ad immunity.
Collapse
Affiliation(s)
- Masahiko Hashimoto
- Department of Genetic Medicine, Weill Medical College of Cornell University, New York, NY 10021, USA
| | | | | | | | | |
Collapse
|
37
|
Bangari DS, Mittal SK. Development of nonhuman adenoviruses as vaccine vectors. Vaccine 2005; 24:849-62. [PMID: 16297508 PMCID: PMC1462960 DOI: 10.1016/j.vaccine.2005.08.101] [Citation(s) in RCA: 108] [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: 05/19/2005] [Revised: 07/29/2005] [Accepted: 08/25/2005] [Indexed: 12/30/2022]
Abstract
Human adenoviral (HAd) vectors have demonstrated great potential as vaccine vectors. Preclinical and clinical studies have demonstrated the feasibility of vector design, robust antigen expression and protective immunity using this system. However, clinical use of adenoviral vectors for vaccine purposes is anticipated to be limited by vector immunity that is either preexisting or develops rapidly following the first inoculation with adenoviral vectors. Vector immunity inactivates the vector particles and rapidly removes the transduced cells, thereby limiting the duration of transgene expression. Due to strong vector immunity, subsequent use of the same vector is usually less efficient. In order to circumvent this limitation, nonhuman adenoviral vectors have been proposed as alternative vectors. In addition to eluding HAd immunity, these vectors possess most of the attractive features of HAd vectors. Several replication-competent or replication-defective nonhuman adenoviral vectors have been developed and investigated for their potential as vaccine-delivery vectors. Here, we review recent advances in the design and characterization of various nonhuman adenoviral vectors, and discuss their potential applications for human and animal vaccination.
Collapse
Affiliation(s)
| | - Suresh K. Mittal
- Corresponding author. Tel.: +1 765 496 2894; fax: +1 765 494 9830.
| |
Collapse
|
38
|
Zeng M, Xu Q, Hesek ED, Pichichero ME. N-fragment of edema factor as a candidate antigen for immunization against anthrax. Vaccine 2005; 24:662-70. [PMID: 16157430 DOI: 10.1016/j.vaccine.2005.08.056] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2005] [Revised: 06/27/2005] [Accepted: 08/05/2005] [Indexed: 11/21/2022]
Abstract
The nontoxic N-terminal fragment of Bacillus anthracis edema factor (EF) was evaluated as a candidate antigen in an anthrax vaccine using a replication-incompetent adenoviral vector. An E1/E3 deleted adenovirus (Ad/EFn) encoding the N-terminal region 1-254 amino acids of the edema factor (EFn) was constructed using the native DNA sequence of EFn. Intramuscular immunization three times with 10(8) plaque forming units (pfu)/dose of Ad/EFn in A/J mice resulted in 37% and 57% protection against a subcutaneous challenge with B. anthracis Sterne strain spores at a dosage of 200 x LD50 and 100 x LD50, respectively. EF-specific serum IgG responses (including total IgG, IgG1, and IgG2a isotype titers) were robust in the Ad/EFn immunized animals. Interestingly, anti-EF antibodies cross-reacted with anthrax lethal factor (LF), and had a neutralizing capability against both anthrax lethal toxin (Letx) and edema toxin (Edtx), as demonstrated by in vitro toxin neutralization assays using J774A.1 mouse macrophage and Chinese hamster ovary cell (CHO), respectively. Our data suggest that EF plays a role in eliciting protective immunity against anthrax, and that it should be included in a new generation multi-component subunit vaccine.
Collapse
Affiliation(s)
- Mingtao Zeng
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Box 672, Rochester, NY 14642, USA.
| | | | | | | |
Collapse
|
39
|
Lemckert AAC, Sumida SM, Holterman L, Vogels R, Truitt DM, Lynch DM, Nanda A, Ewald BA, Gorgone DA, Lifton MA, Goudsmit J, Havenga MJE, Barouch DH. Immunogenicity of heterologous prime-boost regimens involving recombinant adenovirus serotype 11 (Ad11) and Ad35 vaccine vectors in the presence of anti-ad5 immunity. J Virol 2005; 79:9694-701. [PMID: 16014931 PMCID: PMC1181575 DOI: 10.1128/jvi.79.15.9694-9701.2005] [Citation(s) in RCA: 134] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The high prevalence of preexisting immunity to adenovirus serotype 5 (Ad5) in human populations will likely limit the immunogenicity and clinical utility of recombinant Ad5 (rAd5) vector-based vaccines for human immunodeficiency virus type 1 and other pathogens. A potential solution to this problem is to utilize rAd vaccine vectors derived from rare Ad serotypes such as Ad35 and Ad11. We have previously reported that rAd35 vectors were immunogenic in the presence of anti-Ad5 immunity, but the immunogenicity of heterologous rAd prime-boost regimens and the extent that cross-reactive anti-vector immunity may limit this approach have not been fully explored. Here we assess the immunogenicity of heterologous vaccine regimens involving rAd5, rAd35, and novel rAd11 vectors expressing simian immunodeficiency virus Gag in mice both with and without anti-Ad5 immunity. Heterologous rAd prime-boost regimens proved significantly more immunogenic than homologous regimens, as expected. Importantly, all regimens that included rAd5 were markedly suppressed by anti-Ad5 immunity. In contrast, rAd35-rAd11 and rAd11-rAd35 regimens elicited high-frequency immune responses both in the presence and in the absence of anti-Ad5 immunity, although we also detected clear cross-reactive Ad35/Ad11-specific humoral and cellular immune responses. Nevertheless, these data suggest the potential utility of heterologous rAd prime-boost vaccine regimens using vectors derived from rare human Ad serotypes.
Collapse
|
40
|
Wedemeyer J, Malek NP, Manns MP, Bahr MJ. Molekulare Therapie in der Gastroenterologie und Hepatologie. Internist (Berl) 2005; 46:861-2, 864-8, 870-2. [PMID: 15997383 DOI: 10.1007/s00108-005-1462-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
During recent years, molecular techniques have significantly impacted our understanding and therapeutic concepts in gastrointestinal and liver disease. In a number of diseases, diagnostic work-up includes molecular data that supplements the phenotypical evaluation. This includes monogenic diseases as well as the identification of genetic risk factors (e. g. NOD2/CARD15 mutation in Crohn's disease) and viral disease. Attempts to replace liver transplantation in hereditary liver disease by targeted molecular interventions (e. g. via viral vectors) are still experimental, but the associated techniques have improved considerably. The molecular identification of therapeutic targets was followed by the development of specifically tailored therapeutics. These agents are mainly used in the treatment of chronic inflammatory bowel disease and gastrointestinal tumors.
Collapse
Affiliation(s)
- J Wedemeyer
- Abteilung Gastroenterologie, Hepatologie und Endokrinologie, Medizinische Hochschule Hannover, Germany
| | | | | | | |
Collapse
|
41
|
Bangari DS, Sharma A, Mittal SK. Bovine adenovirus type 3 internalization is independent of primary receptors of human adenovirus type 5 and porcine adenovirus type 3. Biochem Biophys Res Commun 2005; 331:1478-84. [PMID: 15883040 PMCID: PMC1552094 DOI: 10.1016/j.bbrc.2005.04.058] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2005] [Indexed: 10/25/2022]
Abstract
Usefulness of adenoviral vectors derived from human adenovirus (HAd) type 5 (HAd5) is mainly limited by wide prevalence of preexisting anti-HAd5 immunity as well as non-specific tissue tropism of these vectors. As an alternative, non-human adenoviral vectors including bovine adenovirus type 3 (BAd3) are currently being investigated. Non-prevalence of BAd3 in humans and its ability to evade preexisting HAd immunity are some of the features that make BAd3 a promising vector for human gene delivery. BAd3 appears to have a tissue tropism distinct from that of HAd5 and also the repertoire of cells efficiently transduced by BAd3 is different. We performed antibody-mediated receptor blocking experiments to show that BAd3 internalization was independent of coxsackievirus-adenovirus receptor, the primary determinant of HAd5 tropism, or integrin alpha(v)beta3, a secondary molecule involved in HAd5 entry. Using homologous and heterologous knob-mediated competition assays with recombinant knobs of HAd5, porcine adenovirus type 3 (PAd3), or BAd3, we observed that BAd3 internalization was independent of the primary receptors of HAd5 and PAd3. These results provide support for further exploration of BAd3 vectors for designing targeted vectors for human gene therapy.
Collapse
Affiliation(s)
- Dinesh S Bangari
- Laboratory of Gene Therapy, Department of Pathobiology and Purdue University Cancer Center, Purdue University, West Lafayette, IN 47907, USA
| | | | | |
Collapse
|
42
|
Stone D, Ni S, Li ZY, Gaggar A, DiPaolo N, Feng Q, Sandig V, Lieber A. Development and assessment of human adenovirus type 11 as a gene transfer vector. J Virol 2005; 79:5090-104. [PMID: 15795294 PMCID: PMC1069572 DOI: 10.1128/jvi.79.8.5090-5104.2005] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Adenovirus vectors based on human serotype 5 (Ad5) have successfully been used as gene transfer vectors in many gene therapy-based approaches to treat disease. Despite their widespread application, many potential therapeutic applications are limited by the widespread prevalence of vector-neutralizing antibodies within the human population and the inability of Ad5-based vectors to transduce important therapeutic target cell types. In an attempt to circumvent these problems, we have developed Ad vectors based on human Ad serotype 11 (Ad11), since the prevalence of neutralizing antibodies to Ad11 in humans is low. E1-deleted Ad11 vector genomes were generated by homologous recombination in 293 cells expressing the Ad11-E1B55K protein or by recombination in Escherichia coli. E1-deleted Ad11 genomes did not display transforming activity in rodent cells. Transduction of primary human CD34+ hematopoietic progenitor cells and immature dendritic cells was more efficient with Ad11 vectors than with Ad5 vectors. Thirty minutes after intravenous injection into mice that express one of the Ad11 receptors (CD46), we found, in a pattern and at a level comparable to what is found in humans, Ad11 vector genomes in all analyzed organs, with the highest amounts in liver, lung, kidney, and spleen. Neither Ad11 genomes nor Ad11 vector-mediated transgene expression were, however, detected at 72 h postinfusion. A large number of Ad11 particles were also found to be associated with circulating blood cells. We also discovered differences in in vitro transduction efficiencies and in vivo biodistributions between Ad11 vectors and chimeric Ad5 vectors possessing Ad11 fibers, indicating that Ad11 capsid proteins other than fibers influence viral infectivity and tropism. Overall, our study provides a basis for the application of Ad11 vectors for in vitro and in vivo gene transfer and for gaining an understanding of the factors that determine Ad tropism.
Collapse
Affiliation(s)
- Daniel Stone
- University of Washington, Division of Medical Genetics, Box 357720, Seattle, WA 98195, USA
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Bangari DS, Mittal SK. Porcine adenovirus serotype 3 internalization is independent of CAR and alphavbeta3 or alphavbeta5 integrin. Virology 2005; 332:157-66. [PMID: 15661148 DOI: 10.1016/j.virol.2004.11.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2004] [Revised: 10/22/2004] [Accepted: 11/10/2004] [Indexed: 01/09/2023]
Abstract
Nonhuman adenoviruses including porcine adenovirus serotype 3 (PAd3) are emerging vectors for gene delivery. PAd3 efficiently transduces human and murine cells in culture, and circumvents preexisting humoral immunity in humans. The coxsackievirus-adenovirus receptor (CAR) serves as a primary receptor and alphavbeta3 or alphavbeta5 integrin as a secondary receptor for several human adenovirus (HAd) subtypes including HAd5. In this study, we deduced the role of CAR, alphavbeta3 or alphavbeta5 integrin in PAd3 internalization. Transduction experiments were conducted in human mammary epithelial (MCF-10A) cells using replication-defective PAd-GFP (PAd3 vector expressing green fluorescent protein [GFP]) and HAd-GFP (HAd5 vector expressing GFP). MCF-10A cells were treated with or without anti-human CAR, or anti-alphavbeta3 or anti-alphavbeta5 integrin antibodies prior to infection with HAd-GFP or PAd-GFP. Significant (P <0.05) inhibition in transduction by HAd-GFP was observed in antibody-treated cells as compared to untreated cells, whereas transduction by PAd-GFP remained to similar levels irrespective of the treatment. To study the adenoviral fiber knob-mediated virus interference, MCF-10A cells were treated with or without the recombinant HAd5 or PAd3 knob followed by infection with HAd-GFP or PAd-GFP. Significant (P <0.05) inhibition was observed only in transduction of the homologous vector. These results suggested that PAd3 internalization was CAR- as well as alphavbeta3 or alphavbeta5 integrin-independent and the primary receptor for HAd5 and PAd3 were distinct. CAR- and alphavbeta3 or alphavbeta5 integrin-independent entry of PAd3 vectors may have implications in targeting cell types that are not efficiently transduced by other adenoviral vectors.
Collapse
MESH Headings
- Adenoviruses, Human/genetics
- Adenoviruses, Human/immunology
- Adenoviruses, Human/physiology
- Adenoviruses, Porcine/genetics
- Adenoviruses, Porcine/immunology
- Adenoviruses, Porcine/physiology
- Animals
- Endocytosis/physiology
- Genetic Vectors/physiology
- Humans
- Integrin alphaVbeta3/physiology
- Integrins/physiology
- Receptors, Virus/metabolism
- Receptors, Vitronectin/physiology
- Transduction, Genetic/methods
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Dinesh S Bangari
- Laboratory of Gene Therapy, Department of Veterinary Pathobiology, and the Purdue Cancer Center, Purdue University, West Lafayette, IN 47907, USA
| | | |
Collapse
|
44
|
Abstract
Adenoviruses have transitioned from tools for gene replacement therapy to bona fide vaccine delivery vehicles. They are attractive vaccine vectors as they induce both innate and adaptive immune responses in mammalian hosts. Currently, adenovirus vectors are being tested as subunit vaccine systems for numerous infectious agents ranging from malaria to HIV-1. Additionally, they are being explored as vaccines against a multitude of tumor-associated antigens. In this review we describe the molecular biology of adenoviruses as well as ways the adenovirus vectors can be manipulated to enhance their efficacy as vaccine carriers. We describe methods of evaluating immune responses to transgene products expressed by adenoviral vectors and discuss data on adenoviral vaccines to a selected number of pathogens. Last, we comment on the limitations of using human adenoviral vectors and provide alternatives to circumvent these problems. This field is growing at an exciting and rapid pace, thus we have limited our scope to the use of adenoviral vectors as vaccines against viral pathogens.
Collapse
Affiliation(s)
| | - Hildegund C.J. Ertl
- To whom correspondence and reprint requests should be addressed. Fax: +1 (215) 898 3953
| |
Collapse
|
45
|
Bangari DS, Shukla S, Mittal SK. Comparative transduction efficiencies of human and nonhuman adenoviral vectors in human, murine, bovine, and porcine cells in culture. Biochem Biophys Res Commun 2005; 327:960-6. [PMID: 15649439 DOI: 10.1016/j.bbrc.2004.12.099] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2004] [Indexed: 11/18/2022]
Abstract
Clinical usefulness of human Ad serotype 5 (HAd5) based vectors is limited primarily because of preexisting Ad immunity and lack of targeting to specific cell types. Alternative vectors based on less prevalent HAd serotypes as well as nonhuman adenoviruses such as porcine Ad serotype 3 (PAd3) and bovine Ad serotype 3 (BAd3) are being developed to overcome these shortcomings. Using virus neutralization assay, we examined whether preexisting Ad immunity in humans would cross-neutralize PAd3 or BAd3. To further evaluate the potential of PAd3 and BAd3 vectors as gene delivery vehicles, we compared their transduction efficiencies in a panel of human, murine, bovine, and porcine cell lines to those obtained with a HAd5 vector. Transduction by the HAd5 vector in the majority of human cell lines correlated with the expression levels of coxsackievirus-adenovirus receptor (CAR), the primary HAd5 receptor; while transduction by PAd3 and BAd3 vectors was CAR-independent. The results suggest that PAd3 and BAd3 vectors are promising gene delivery vehicles for human gene therapy as well as for recombinant vaccines for human and animal use.
Collapse
Affiliation(s)
- Dinesh S Bangari
- Laboratory of Gene Therapy, Department of Pathobiology, and Purdue University Cancer Center, Purdue University, West Lafayette, IN 47907, USA
| | | | | |
Collapse
|
46
|
|
47
|
|
48
|
Barouch DH, Nabel GJ. Adenovirus Vector-Based Vaccines for Human Immunodeficiency Virus Type 1. Hum Gene Ther 2005; 16:149-56. [PMID: 15761255 DOI: 10.1089/hum.2005.16.149] [Citation(s) in RCA: 142] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Recombinant adenovirus (rAd) vectors have received considerable attention for gene therapy because of their high transduction efficiency. However, recombinant gene expression from rAd vectors elicits rapid and potent immune responses to foreign transgene products. Such immunogenicity limits the duration of transgene expression and poses a major challenge to the use of rAd vectors for gene therapy. In contrast, the inherent immunogenicity of these vectors is a desirable feature for vaccine development. The immunogenicity and protective efficacy of rAd vector-based vaccines have now been demonstrated in a number of animal models, and rAd vaccines for a variety of pathogens are currently being explored in early-phase clinical trials. In this review, we describe progress in the development of rAd vector-based vaccines with a focus on human immunodeficiency virus type 1.
Collapse
Affiliation(s)
- Dan H Barouch
- Division of Viral Pathogenesis, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA.
| | | |
Collapse
|
49
|
Wang XY, Martiniello-Wilks R, Shaw JM, Ho T, Coulston N, Cooke-Yarborough C, Molloy PL, Cameron F, Moghaddam M, Lockett TJ, Webster LK, Smith IK, Both GW, Russell PJ. Preclinical evaluation of a prostate-targeted gene-directed enzyme prodrug therapy delivered by ovine atadenovirus. Gene Ther 2005; 11:1559-67. [PMID: 15343359 DOI: 10.1038/sj.gt.3302308] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Gene-directed enzyme prodrug therapy (GDEPT) based on the Escherichia coli enzyme, purine nucleoside phosphorylase (PNP), provides a novel strategy for treating slowly growing tumors like prostate cancer (CaP). PNP converts systemically administered prodrug, fludarabine phosphate, to a toxic metabolite, 2-fluoroadenine, that kills PNP-expressing and nearby cells by inhibiting DNA, RNA and protein synthesis. Reporter gene expression directed by a hybrid prostate-directed promoter and enhancer, PSMEPb, was assayed after plasmid transfection or viral transduction of prostate and non-CaP cell lines. Androgen-sensitive (AS) LNCaP-LN3 and androgen-independent (AI) PC3 human CaP xenografts in nude mice were injected intratumorally with an ovine atadenovirus vector, OAdV623, that carries the PNP gene under PSMEPb, formulated with cationic lipid for enhanced infectivity. Fludarabine phosphate was then given intraperitoneally for 5 days at 75 mg/m2/day. PNP expression was evaluated by enzymic conversion of its substrate using reverse phase HPLC. OAdV623 showed excellent in vitro transcriptional specificity for CaP cells. In vivo, expression of PNP persisted for > 6 days after OAdV623 injection and a single treatment provided 100% increase in tumor doubling time and > 50% inhibition of tumor growth for both LNCaP-LN3 and PC3 lines, with increased tumor necrosis and apoptosis and decreased tumor cell proliferation. OAdV623 significantly suppressed the growth of AS and AI human CaP xenografts in mice.
Collapse
Affiliation(s)
- X Y Wang
- Oncology Research Centre, Prince of Wales Hospital Clinical School of Medicine, The University of New South Wales, Randwick, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Rosenthal KL. Recombinant Live Viral Vectors as Vaccines for Mucosal Immunity. Mucosal Immunol 2005. [DOI: 10.1016/b978-012491543-5/50061-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|