1
|
Santos LC, Fernandes AMS, Alves IA, Serafini MR, Silva LDSE, de Freitas HF, Leite LCC, Santos CC. Trends in Viral Vector-Based Vaccines for Tuberculosis: A Patent Review (2010-2023). Vaccines (Basel) 2024; 12:876. [PMID: 39204002 PMCID: PMC11359462 DOI: 10.3390/vaccines12080876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 07/25/2024] [Accepted: 07/30/2024] [Indexed: 09/03/2024] Open
Abstract
Tuberculosis (TB) is an ancient global public health problem. Several strategies have been applied to develop new and more effective vaccines against TB, from attenuated or inactivated mycobacteria to recombinant subunit or genetic vaccines, including viral vectors. This review aimed to evaluate patents filed between 2010 and 2023 for TB vaccine candidates. It focuses on viral vector-based strategies. A search was carried out in Espacenet, using the descriptors "mycobacterium and tuberculosis" and the classification A61K39. Of the 411 patents preliminarily identified, the majority were related to subunit vaccines, with 10 patents based on viral vector platforms selected in this study. Most of the identified patents belong to the United States or China, with a concentration of patent filings between 2013 and 2023. Adenoviruses were the most explored viral vectors, and the most common immunodominant Mycobacterium tuberculosis (Mtb) antigens were present in all the selected patents. The majority of patents were tested in mouse models by intranasal or subcutaneous route of immunization. In the coming years, an increased use of this platform for prophylactic and/or therapeutic approaches for TB and other diseases is expected. Along with this, expanding knowledge about the safety of this technology is essential to advance its use.
Collapse
Affiliation(s)
- Lana C. Santos
- Serviço de Imunologia das Doenças Infecciosas, Faculdade de Farmácia, Universidade Federal da Bahia, Salvador 40170-115, BA, Brazil; (L.C.S.); (A.M.S.F.); (L.d.S.e.S.)
| | - Antônio Márcio Santana Fernandes
- Serviço de Imunologia das Doenças Infecciosas, Faculdade de Farmácia, Universidade Federal da Bahia, Salvador 40170-115, BA, Brazil; (L.C.S.); (A.M.S.F.); (L.d.S.e.S.)
| | - Izabel Almeida Alves
- Departamento do Medicamento, Faculdade de Farmácia, Universidade Federal da Bahia, Salvador 40170-115, BA, Brazil;
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade do Estado da Bahia, Salvador 41150-000, BA, Brazil
| | - Mairim Russo Serafini
- Departamento de Farmácia, Universidade Federal do Sergipe, São Cristóvão 49100-000, SE, Brazil;
| | - Leandra da Silva e Silva
- Serviço de Imunologia das Doenças Infecciosas, Faculdade de Farmácia, Universidade Federal da Bahia, Salvador 40170-115, BA, Brazil; (L.C.S.); (A.M.S.F.); (L.d.S.e.S.)
| | | | - Luciana C. C. Leite
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo 05503-900, SP, Brazil;
| | - Carina C. Santos
- Serviço de Imunologia das Doenças Infecciosas, Faculdade de Farmácia, Universidade Federal da Bahia, Salvador 40170-115, BA, Brazil; (L.C.S.); (A.M.S.F.); (L.d.S.e.S.)
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal da Bahia, Salvador 40170-115, BA, Brazil
| |
Collapse
|
2
|
Sayedahmed EE, Elshafie NO, Zhang G, Mohammed SI, Sambhara S, Mittal SK. Enhancement of mucosal innate and adaptive immunity following intranasal immunization of mice with a bovine adenoviral vector. Front Immunol 2023; 14:1305937. [PMID: 38077380 PMCID: PMC10702558 DOI: 10.3389/fimmu.2023.1305937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 11/01/2023] [Indexed: 12/18/2023] Open
Abstract
Introduction Nonhuman adenoviral (AdV) gene delivery platforms have significant value due to their ability to elude preexisting AdV vector immunity in most individuals. Previously, we have demonstrated that intranasal (IN) immunization of mice with BAd-H5HA, a bovine AdV type 3 (BAdV3) vector expressing H5N1 influenza virus hemagglutinin (HA), resulted in enhanced humoral and cell-mediated immune responses. The BAd-H5HA IN immunization resulted in complete protection following the challenge with an antigenically distinct H5N1 virus compared to the mouse group similarly immunized with HAd-H5HA, a human AdV type 5 (HAdV5) vector expressing HA. Methods Here, we attempted to determine the activation of innate immune responses in the lungs of mice inoculated intranasally with BAd-H5HA compared to the HAd-H5HA-inoculated group. Results RNA-Seq analyses of the lung tissues revealed differential expression (DE) of genes involved in innate and adaptive immunity in animals immunized with BAd-H5HA. The top ten enhanced genes were verified by RT-PCR. Consistently, there were transient increases in the levels of cytokines (IL-1α, IL-1β, IL-5, TNF- α, LIF, IL-17, G-CSF, MIP-1β, MCP-1, MIP-2, and GM-CSF) and toll-like receptors in the lungs of the group inoculated with BAdV vectors compared to that of the HAdV vector group. Conclusion These results demonstrate that the BAdV vectors induce enhanced innate and adaptive immunity-related factors compared to HAdV vectors in mice. Thus, the BAdV vector platform could be an excellent gene delivery system for recombinant vaccines and cancer immunotherapy.
Collapse
Affiliation(s)
- Ekramy E. Sayedahmed
- Department of Comparative Pathobiology, Purdue Institute for Immunology, Inflammation and Infectious Diseases, and Purdue University Center for Cancer Research, College of Veterinary Medicine, Purdue University, West Lafayette, IN, United States
| | - Nelly O. Elshafie
- Department of Comparative Pathobiology, Purdue Institute for Immunology, Inflammation and Infectious Diseases, and Purdue University Center for Cancer Research, College of Veterinary Medicine, Purdue University, West Lafayette, IN, United States
| | - GuangJun Zhang
- Department of Comparative Pathobiology, Purdue Institute for Immunology, Inflammation and Infectious Diseases, and Purdue University Center for Cancer Research, College of Veterinary Medicine, Purdue University, West Lafayette, IN, United States
| | - Sulma I. Mohammed
- Department of Comparative Pathobiology, Purdue Institute for Immunology, Inflammation and Infectious Diseases, and Purdue University Center for Cancer Research, College of Veterinary Medicine, Purdue University, West Lafayette, IN, United States
| | - Suryaprakash Sambhara
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Suresh K. Mittal
- Department of Comparative Pathobiology, Purdue Institute for Immunology, Inflammation and Infectious Diseases, and Purdue University Center for Cancer Research, College of Veterinary Medicine, Purdue University, West Lafayette, IN, United States
| |
Collapse
|
3
|
Sayedahmed EE, Araújo MV, Silva-Pereira TT, Chothe SK, Elkashif A, Alhashimi M, Wang WC, Santos AP, Nair MS, Gontu A, Nissly R, Francisco de Souza Filho A, Tavares MS, Ayupe MC, Salgado CL, Donizetti de Oliveira Candido É, Leal Oliveira DB, Durigon EL, Heinemann MB, Morais da Fonseca D, Jagannath C, Sá Guimarães AM, Kuchipudi SV, Mittal SK. Impact of an autophagy-inducing peptide on immunogenicity and protection efficacy of an adenovirus-vectored SARS-CoV-2 vaccine. Mol Ther Methods Clin Dev 2023; 30:194-207. [PMID: 37502665 PMCID: PMC10299838 DOI: 10.1016/j.omtm.2023.06.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 06/23/2023] [Indexed: 07/29/2023]
Abstract
Because of continual generation of new variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), it is critical to design the next generation of vaccines to combat the threat posed by SARS-CoV-2 variants. We developed human adenovirus (HAd) vector-based vaccines (HAd-Spike/C5 and HAd-Spike) that express the whole Spike (S) protein of SARS-CoV-2 with or without autophagy-inducing peptide C5 (AIP-C5), respectively. Mice or golden Syrian hamsters immunized intranasally (i.n.) with HAd-Spike/C5 induced similar levels of S-specific humoral immune responses and significantly higher levels of S-specific cell-mediated immune (CMI) responses compared with HAd-Spike vaccinated groups. These results indicated that inclusion of AIP-C5 induced enhanced S-specific CMI responses and similar levels of virus-neutralizing titers against SARS-CoV-2 variants. To investigate the protection efficacy, golden Syrian hamsters immunized i.n. either with HAd-Spike/C5 or HAd-Spike were challenged with SARS-CoV-2. The lungs and nasal turbinates were collected 3, 5, 7, and 14 days post challenge. Significant reductions in morbidity, virus titers, and lung histopathological scores were observed in immunized groups compared with the mock- or empty vector-inoculated groups. Overall, slightly better protection was seen in the HAd-Spike/C5 group compared with the HAd-Spike group.
Collapse
Affiliation(s)
- Ekramy E. Sayedahmed
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
- Purdue Institute of Inflammation, Immunology, and Infectious Disease, Purdue University, West Lafayette, IN, USA
- Institute for Cancer Research, Purdue University, West Lafayette, IN, USA
| | - Marcelo Valdemir Araújo
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Butantan Institute, São Paulo, Brazil
| | - Taiana Tainá Silva-Pereira
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Shubhada K. Chothe
- Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, PA, USA
| | - Ahmed Elkashif
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
- Purdue Institute of Inflammation, Immunology, and Infectious Disease, Purdue University, West Lafayette, IN, USA
- Institute for Cancer Research, Purdue University, West Lafayette, IN, USA
| | - Marwa Alhashimi
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
- Purdue Institute of Inflammation, Immunology, and Infectious Disease, Purdue University, West Lafayette, IN, USA
- Institute for Cancer Research, Purdue University, West Lafayette, IN, USA
| | - Wen-Chien Wang
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
- Purdue Institute of Inflammation, Immunology, and Infectious Disease, Purdue University, West Lafayette, IN, USA
- Institute for Cancer Research, Purdue University, West Lafayette, IN, USA
| | - Andrea P. Santos
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
- Purdue Institute of Inflammation, Immunology, and Infectious Disease, Purdue University, West Lafayette, IN, USA
- Institute for Cancer Research, Purdue University, West Lafayette, IN, USA
| | - Meera Surendran Nair
- Department of Veterinary and Biomedical Sciences, Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, USA
- Animal Diagnostic Laboratory, Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, PA, USA
| | - Abhinay Gontu
- Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, PA, USA
| | - Ruth Nissly
- Animal Diagnostic Laboratory, Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, PA, USA
| | | | - Mariana Silva Tavares
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Marina Caçador Ayupe
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Caio Loureiro Salgado
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | | | | - Edison Luiz Durigon
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Marcos Bryan Heinemann
- Department of Preventive Veterinary Medicine and Animal Health, College of Veterinary Medicine, University of São Paulo, São Paulo, Brazil
| | - Denise Morais da Fonseca
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Chinnaswamy Jagannath
- Department of Pathology and Genomic Medicine, Center for Infectious Diseases and Translational Medicine, Houston Methodist Research Institute, Houston, TX, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Ana Marcia Sá Guimarães
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Suresh V. Kuchipudi
- Animal Diagnostic Laboratory, Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, PA, USA
| | - Suresh K. Mittal
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
- Purdue Institute of Inflammation, Immunology, and Infectious Disease, Purdue University, West Lafayette, IN, USA
- Institute for Cancer Research, Purdue University, West Lafayette, IN, USA
| |
Collapse
|
4
|
Cordeiro AS, Patil-Sen Y, Shivkumar M, Patel R, Khedr A, Elsawy MA. Nanovaccine Delivery Approaches and Advanced Delivery Systems for the Prevention of Viral Infections: From Development to Clinical Application. Pharmaceutics 2021; 13:2091. [PMID: 34959372 PMCID: PMC8707864 DOI: 10.3390/pharmaceutics13122091] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 11/30/2021] [Accepted: 12/01/2021] [Indexed: 02/07/2023] Open
Abstract
Viral infections causing pandemics and chronic diseases are the main culprits implicated in devastating global clinical and socioeconomic impacts, as clearly manifested during the current COVID-19 pandemic. Immunoprophylaxis via mass immunisation with vaccines has been shown to be an efficient strategy to control such viral infections, with the successful and recently accelerated development of different types of vaccines, thanks to the advanced biotechnological techniques involved in the upstream and downstream processing of these products. However, there is still much work to be done for the improvement of efficacy and safety when it comes to the choice of delivery systems, formulations, dosage form and route of administration, which are not only crucial for immunisation effectiveness, but also for vaccine stability, dose frequency, patient convenience and logistics for mass immunisation. In this review, we discuss the main vaccine delivery systems and associated challenges, as well as the recent success in developing nanomaterials-based and advanced delivery systems to tackle these challenges. Manufacturing and regulatory requirements for the development of these systems for successful clinical and marketing authorisation were also considered. Here, we comprehensively review nanovaccines from development to clinical application, which will be relevant to vaccine developers, regulators, and clinicians.
Collapse
Affiliation(s)
- Ana Sara Cordeiro
- Leicester Institute for Pharmaceutical Innovation, Leicester School of Pharmacy, De Montfort University, Leicester LE1 9BH, UK; (A.S.C.); (M.S.); (A.K.)
| | - Yogita Patil-Sen
- Wrightington, Wigan and Leigh Teaching Hospitals NHS Foundation Trust, National Health Service, Wigan WN6 0SZ, UK;
| | - Maitreyi Shivkumar
- Leicester Institute for Pharmaceutical Innovation, Leicester School of Pharmacy, De Montfort University, Leicester LE1 9BH, UK; (A.S.C.); (M.S.); (A.K.)
| | - Ronak Patel
- School of Pharmacy and Biomedical Sciences, University of Central Lancashire, Preston PR1 2HE, UK;
| | - Abdulwahhab Khedr
- Leicester Institute for Pharmaceutical Innovation, Leicester School of Pharmacy, De Montfort University, Leicester LE1 9BH, UK; (A.S.C.); (M.S.); (A.K.)
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Mohamed A. Elsawy
- Leicester Institute for Pharmaceutical Innovation, Leicester School of Pharmacy, De Montfort University, Leicester LE1 9BH, UK; (A.S.C.); (M.S.); (A.K.)
| |
Collapse
|
5
|
Elkashif A, Alhashimi M, Sayedahmed EE, Sambhara S, Mittal SK. Adenoviral vector-based platforms for developing effective vaccines to combat respiratory viral infections. Clin Transl Immunology 2021; 10:e1345. [PMID: 34667600 PMCID: PMC8510854 DOI: 10.1002/cti2.1345] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 09/10/2021] [Accepted: 09/15/2021] [Indexed: 02/06/2023] Open
Abstract
Since the development of the first vaccine against smallpox over two centuries ago, vaccination strategies have been at the forefront of significantly impacting the incidences of infectious diseases globally. However, the increase in the human population, deforestation and climate change, and the rise in worldwide travel have favored the emergence of new viruses with the potential to cause pandemics. The ongoing severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic is a cruel reminder of the impact of novel pathogens and the suboptimal capabilities of conventional vaccines. Therefore, there is an urgent need to develop new vaccine strategies that allow the production of billions of doses in a short duration and are broadly protective against emerging and re-emerging infectious diseases. Extensive knowledge of the molecular biology and immunology of adenoviruses (Ad) has favored Ad vectors as platforms for vaccine design. The Ad-based vaccine platform represents an attractive strategy as it induces robust humoral and cell-mediated immune responses and can meet the global demand in a pandemic situation. This review describes the status of Ad vector-based vaccines in preclinical and clinical studies for current and emerging respiratory viruses, particularly coronaviruses, influenza viruses and respiratory syncytial viruses.
Collapse
Affiliation(s)
- Ahmed Elkashif
- Department of Comparative PathobiologyPurdue Institute for Inflammation, Immunology and Infectious Disease, and Purdue University Center for Cancer ResearchCollege of Veterinary MedicinePurdue UniversityWest LafayetteINUSA
| | - Marwa Alhashimi
- Department of Comparative PathobiologyPurdue Institute for Inflammation, Immunology and Infectious Disease, and Purdue University Center for Cancer ResearchCollege of Veterinary MedicinePurdue UniversityWest LafayetteINUSA
| | - Ekramy E Sayedahmed
- Department of Comparative PathobiologyPurdue Institute for Inflammation, Immunology and Infectious Disease, and Purdue University Center for Cancer ResearchCollege of Veterinary MedicinePurdue UniversityWest LafayetteINUSA
| | | | - Suresh K Mittal
- Department of Comparative PathobiologyPurdue Institute for Inflammation, Immunology and Infectious Disease, and Purdue University Center for Cancer ResearchCollege of Veterinary MedicinePurdue UniversityWest LafayetteINUSA
| |
Collapse
|
6
|
Khan A, Sayedahmed EE, Singh VK, Mishra A, Dorta-Estremera S, Nookala S, Canaday DH, Chen M, Wang J, Sastry KJ, Mittal SK, Jagannath C. A recombinant bovine adenoviral mucosal vaccine expressing mycobacterial antigen-85B generates robust protection against tuberculosis in mice. Cell Rep Med 2021; 2:100372. [PMID: 34467249 PMCID: PMC8385328 DOI: 10.1016/j.xcrm.2021.100372] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 02/16/2021] [Accepted: 07/19/2021] [Indexed: 02/07/2023]
Abstract
Although the BCG vaccine offers partial protection, tuberculosis remains a leading cause of infectious disease death, killing ∼1.5 million people annually. We developed mucosal vaccines expressing the autophagy-inducing peptide C5 and mycobacterial Ag85B-p25 epitope using replication-defective human adenovirus (HAdv85C5) and bovine adenovirus (BAdv85C5) vectors. BAdv85C5-infected dendritic cells (DCs) expressed a robust transcriptome of genes regulating antigen processing compared to HAdv85C5-infected DCs. BAdv85C5-infected DCs showed enhanced galectin-3/8 and autophagy-dependent in vitro Ag85B-p25 epitope presentation to CD4 T cells. BCG-vaccinated mice were intranasally boosted using HAdv85C5 or BAdv85C5 followed by infection using aerosolized Mycobacterium tuberculosis (Mtb). BAdv85C5 protected mice against tuberculosis both as a booster after BCG vaccine (>1.4-log10 reduction in Mtb lung burden) and as a single intranasal dose (>0.5-log10 reduction). Protection was associated with robust CD4 and CD8 effector (TEM), central memory (TCM), and CD103+/CD69+ lung-resident memory (TRM) T cell expansion, revealing BAdv85C5 as a promising mucosal vaccine for tuberculosis.
Collapse
Affiliation(s)
- Arshad Khan
- Department of Pathology and Genomic Medicine, Houston Methodist Academic Institute, Houston Methodist Research Institute & Weill Cornell Medical College, Houston, TX, USA
| | - Ekramy E. Sayedahmed
- Department of Comparative Pathobiology and Purdue Institute of Inflammation, Immunology, and Infectious Disease, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
| | - Vipul K. Singh
- Department of Pathology and Genomic Medicine, Houston Methodist Academic Institute, Houston Methodist Research Institute & Weill Cornell Medical College, Houston, TX, USA
| | - Abhishek Mishra
- Department of Pathology and Genomic Medicine, Houston Methodist Academic Institute, Houston Methodist Research Institute & Weill Cornell Medical College, Houston, TX, USA
| | | | - Sita Nookala
- Department of Thoracic Head and Neck Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - David H. Canaday
- Department of Medicine, Case Western Reserve University and Cleveland Veterans Affairs, Cleveland, OH, USA
| | - Min Chen
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Jin Wang
- Immunobiology and Transplant Science Center, Houston Methodist Research Institute, and Department of Surgery, Weill Cornell Medical College, Houston, TX, USA
| | - K. Jagannadha Sastry
- Department of Thoracic Head and Neck Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Suresh K. Mittal
- Department of Comparative Pathobiology and Purdue Institute of Inflammation, Immunology, and Infectious Disease, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
| | - Chinnaswamy Jagannath
- Department of Pathology and Genomic Medicine, Houston Methodist Academic Institute, Houston Methodist Research Institute & Weill Cornell Medical College, Houston, TX, USA
| |
Collapse
|
7
|
Alhashimi M, Elkashif A, Sayedahmed EE, Mittal SK. Nonhuman Adenoviral Vector-Based Platforms and Their Utility in Designing Next Generation of Vaccines for Infectious Diseases. Viruses 2021; 13:1493. [PMID: 34452358 PMCID: PMC8402644 DOI: 10.3390/v13081493] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/25/2021] [Accepted: 07/26/2021] [Indexed: 01/01/2023] Open
Abstract
Several human adenoviral (Ad) vectors have been developed for vaccine delivery owing to their numerous advantages, including the feasibility of different vector designs, the robustness of elicited immune responses, safety, and scalability. To expand the repertoire of Ad vectors for receptor usage and circumvention of Ad vector immunity, the use of less prevalent human Ad types or nonhuman Ads were explored for vector design. Notably, many nonhuman Ad vectors have shown great promise in preclinical and clinical studies as vectors for vaccine delivery. This review describes the key features of several nonhuman Ad vector platforms and their implications in developing effective vaccines against infectious diseases.
Collapse
Affiliation(s)
| | | | | | - Suresh K. Mittal
- Immunology and Infectious Disease, and Purdue University Center for Cancer Research, Department of Comparative Pathobiology, Purdue Institute for Inflammation, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907-2027, USA; (M.A.); (A.E.); (E.E.S.)
| |
Collapse
|
8
|
Tournier JN, Kononchik J. Virus Eradication and Synthetic Biology: Changes with SARS-CoV-2? Viruses 2021; 13:569. [PMID: 33800626 PMCID: PMC8066276 DOI: 10.3390/v13040569] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/25/2021] [Accepted: 03/26/2021] [Indexed: 12/24/2022] Open
Abstract
The eradication of infectious diseases has been achieved only once in history, in 1980, with smallpox. Since 1988, significant effort has been made to eliminate poliomyelitis viruses, but eradication is still just out of reach. As the goal of viral disease eradication approaches, the ability to recreate historically eradicated viruses using synthetic biology has the potential to jeopardize the long-term sustainability of eradication. However, the emergence of the severe acute respiratory syndrome-coronavirus (SARS-CoV)-2 pandemic has highlighted our ability to swiftly and resolutely respond to a potential outbreak. This virus has been synthetized faster than any other in the past and is resulting in vaccines before most attenuated candidates reach clinical trials. Here, synthetic biology has the opportunity to demonstrate its truest potential to the public and solidify a footing in the world of vaccines.
Collapse
Affiliation(s)
- Jean-Nicolas Tournier
- Microbiology and Infectious Diseases Department, Institut de Recherche Biomédicale des Armées, 91220 Brétigny-sur-Orge, France;
- CNRS UMR-3569, Innovative Vaccine Laboratory, Virology Department, Institut Pasteur, 75015 Paris, France
- Ecole du Val-de-Grâce, 75005 Paris, France
| | - Joseph Kononchik
- Microbiology and Infectious Diseases Department, Institut de Recherche Biomédicale des Armées, 91220 Brétigny-sur-Orge, France;
- US Army Medical Research Institute of Chemical Defense (USAMRICD), 8350 Ricketts Point Rd., Aberdeen Proving Ground, MD 21010, USA
- Toxicology and Chemical Risk Department, Institut de Recherche Biomédicale des Armées, 91220 Brétigny-sur-Orge, France
| |
Collapse
|
9
|
Sayedahmed EE, Elkashif A, Alhashimi M, Sambhara S, Mittal SK. Adenoviral Vector-Based Vaccine Platforms for Developing the Next Generation of Influenza Vaccines. Vaccines (Basel) 2020; 8:vaccines8040574. [PMID: 33019589 PMCID: PMC7712206 DOI: 10.3390/vaccines8040574] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 09/06/2020] [Accepted: 09/17/2020] [Indexed: 12/20/2022] Open
Abstract
Ever since the discovery of vaccines, many deadly diseases have been contained worldwide, ultimately culminating in the eradication of smallpox and polio, which represented significant medical achievements in human health. However, this does not account for the threat influenza poses on public health. The currently licensed seasonal influenza vaccines primarily confer excellent strain-specific protection. In addition to the seasonal influenza viruses, the emergence and spread of avian influenza pandemic viruses such as H5N1, H7N9, H7N7, and H9N2 to humans have highlighted the urgent need to adopt a new global preparedness for an influenza pandemic. It is vital to explore new strategies for the development of effective vaccines for pandemic and seasonal influenza viruses. The new vaccine approaches should provide durable and broad protection with the capability of large-scale vaccine production within a short time. The adenoviral (Ad) vector-based vaccine platform offers a robust egg-independent production system for manufacturing large numbers of influenza vaccines inexpensively in a short timeframe. In this review, we discuss the progress in the development of Ad vector-based influenza vaccines and their potential in designing a universal influenza vaccine.
Collapse
Affiliation(s)
- Ekramy E. Sayedahmed
- Department of Comparative Pathobiology, Purdue Institute for Immunology, Inflammation and Infectious Disease, Purdue University Center for Cancer Research, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA; (E.E.S.); (A.E.); (M.A.)
| | - Ahmed Elkashif
- Department of Comparative Pathobiology, Purdue Institute for Immunology, Inflammation and Infectious Disease, Purdue University Center for Cancer Research, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA; (E.E.S.); (A.E.); (M.A.)
| | - Marwa Alhashimi
- Department of Comparative Pathobiology, Purdue Institute for Immunology, Inflammation and Infectious Disease, Purdue University Center for Cancer Research, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA; (E.E.S.); (A.E.); (M.A.)
| | - Suryaprakash Sambhara
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA
- Correspondence: (S.S.); (S.K.M.)
| | - Suresh K. Mittal
- Department of Comparative Pathobiology, Purdue Institute for Immunology, Inflammation and Infectious Disease, Purdue University Center for Cancer Research, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA; (E.E.S.); (A.E.); (M.A.)
- Correspondence: (S.S.); (S.K.M.)
| |
Collapse
|
10
|
Allen RJ, Byrnes AP. Interaction of adenovirus with antibodies, complement, and coagulation factors. FEBS Lett 2019; 593:3449-3460. [PMID: 31660588 DOI: 10.1002/1873-3468.13649] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 10/21/2019] [Accepted: 10/23/2019] [Indexed: 12/16/2022]
Abstract
Adenovirus (AdV) is one of the most widely used vectors for gene therapy and vaccine studies due to its excellent transduction efficiency, capacity for large transgenes, and high levels of gene expression. When administered intravascularly, the fate of AdV vectors is heavily influenced by interactions with host plasma proteins. Some plasma proteins can neutralize AdV, but AdV can also specifically bind plasma proteins that protect against neutralization and preserve activity. This review summarizes the plasma proteins that interact with AdV, including antibodies, complement, and vitamin K-dependent coagulation factors. We will also review the complex interactions of these plasma proteins with each other and with cellular proteins, as well as strategies for developing better AdV vectors that evade or manipulate plasma proteins.
Collapse
Affiliation(s)
- Rondine J Allen
- Division of Cellular and Gene Therapies, FDA Center for Biologics Evaluation and Research, Silver Spring, MD, USA
| | - Andrew P Byrnes
- Division of Cellular and Gene Therapies, FDA Center for Biologics Evaluation and Research, Silver Spring, MD, USA
| |
Collapse
|
11
|
Sreenivasan CC, Thomas M, Kaushik RS, Wang D, Li F. Influenza A in Bovine Species: A Narrative Literature Review. Viruses 2019; 11:v11060561. [PMID: 31213032 PMCID: PMC6631717 DOI: 10.3390/v11060561] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 06/10/2019] [Accepted: 06/14/2019] [Indexed: 12/17/2022] Open
Abstract
It is quite intriguing that bovines were largely unaffected by influenza A, even though most of the domesticated and wild animals/birds at the human-animal interface succumbed to infection over the past few decades. Influenza A occurs on a very infrequent basis in bovine species and hence bovines were not considered to be susceptible hosts for influenza until the emergence of influenza D. This review describes a multifaceted chronological review of literature on influenza in cattle which comprises mainly of the natural infections/outbreaks, experimental studies, and pathological and seroepidemiological aspects of influenza A that have occurred in the past. The review also sheds light on the bovine models used in vitro and in vivo for influenza-related studies over recent years. Despite a few natural cases in the mid-twentieth century and seroprevalence of human, swine, and avian influenza viruses in bovines, the evolution and host adaptation of influenza A virus (IAV) in this species suffered a serious hindrance until the novel influenza D virus (IDV) emerged recently in cattle across the world. Supposedly, certain bovine host factors, particularly some serum components and secretory proteins, were reported to have anti-influenza properties, which could be an attributing factor for the resilient nature of bovines to IAV. Further studies are needed to identify the host-specific factors contributing to the differential pathogenetic mechanisms and disease progression of IAV in bovines compared to other susceptible mammalian hosts.
Collapse
Affiliation(s)
- Chithra C Sreenivasan
- Department of Biology and Microbiology, South Dakota State University, Brookings, SD 57007, USA.
| | - Milton Thomas
- Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD 57007, USA.
| | - Radhey S Kaushik
- Department of Biology and Microbiology, South Dakota State University, Brookings, SD 57007, USA.
| | - Dan Wang
- Department of Biology and Microbiology, South Dakota State University, Brookings, SD 57007, USA.
- BioSystems Networks and Translational Research Center (BioSNTR), Brookings, SD 57007, USA.
| | - Feng Li
- Department of Biology and Microbiology, South Dakota State University, Brookings, SD 57007, USA.
- BioSystems Networks and Translational Research Center (BioSNTR), Brookings, SD 57007, USA.
| |
Collapse
|
12
|
Abstract
Various adenovirus (AdV) vector systems have proven to be lucrative options for gene delivery. They can serve as potential vaccine candidates for prevention of several common infectious diseases and hold the promise for gene therapy, especially for cancer. Several AdV vector-based therapies are currently at various stages of clinical trials worldwide, which make an immense interest of both the clinicians and researchers. Since these vectors are easy to manipulate, have broad tropism, and have the capability to yield high titers, this delivery system has a wide range of applications for different clinical settings. This chapter emphasizes on some of the current usages of AdV vectors and their production methods.
Collapse
Affiliation(s)
- Ekramy E Sayedahmed
- Department of Comparative Pathobiology, Purdue Institute for Inflammation, Immunology, and Infectious Disease, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
| | - Rashmi Kumari
- Department of Comparative Pathobiology, Purdue Institute for Inflammation, Immunology, and Infectious Disease, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
| | - Suresh K Mittal
- Department of Comparative Pathobiology, Purdue Institute for Inflammation, Immunology, and Infectious Disease, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA.
| |
Collapse
|
13
|
Longevity of adenovirus vector immunity in mice and its implications for vaccine efficacy. Vaccine 2018; 36:6744-6751. [PMID: 30266488 DOI: 10.1016/j.vaccine.2018.09.031] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 09/13/2018] [Accepted: 09/16/2018] [Indexed: 12/18/2022]
Abstract
There is a high incidence of adenovirus (AdV) infection in humans due to the presence of more than 60 types of human adenoviruses (HAdVs). The majority of individuals are exposed to one or more HAdV types early in their lives, leading to the development of AdV type-specific neutralizing antibodies. Similarly, immunization or gene therapy with AdV vectors leads to immune responses to the AdV vector. This 'vector immunity' is a concern for AdV vector-based applications for vaccines or gene therapy, especially when the repeated administration of a vector is required. The objective of this investigation was to establish whether AdV neutralizing antibody titers decline sufficiently in a year to permit annual vaccination with the same AdV vector. Naïve or human adenoviral vector group C, type 5 (HAdV-C5)-primed mice were mock-inoculated (with PBS) or inoculated i.m. with 108 PFU of either HAd-GFP [HAdV-C5 vector expressing the green fluorescent protein (GFP)] to mimic the conditions for the first inoculation with an AdV vector-based vaccine. At 1, 3, 6, and 10 months post-HAd-GFP inoculation, naïve- or HAdV-primed animals were vaccinated i.m. with 108 PFU of HAd-H5HA [HAdV-C5 vector expressing hemagglutinin (HA) of H5N1 influenza virus]. There was a significant continual decrease in vector immunity titers with time, thereby leading to significant continual increases in the levels of HA-specific humoral and cell-mediated immune responses. In addition, significant improvement in protection efficacy against challenge with an antigenically heterologous H5N1 virus was observed in HAdV-primed animals at 6 months and onwards. These results indicate that the annual immunization with the same AdV vector may be effective due to a significant decline in vector immunity.
Collapse
|
14
|
A Bovine Adenoviral Vector-Based H5N1 Influenza -Vaccine Provides Enhanced Immunogenicity and Protection at a Significantly Low Dose. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2018; 10:210-222. [PMID: 30101154 PMCID: PMC6082999 DOI: 10.1016/j.omtm.2018.07.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 07/11/2018] [Indexed: 11/26/2022]
Abstract
Several human and nonhuman adenovirus (AdV) vectors including bovine AdV type 3 (BAdV-3) were developed as gene delivery vectors to supplement and/or elude human AdV (HAdV)-specific neutralizing antibodies (vector immunity). Here we evaluated the vaccine immunogenicity and efficacy of BAdV-3 vector (BAd-H5HA) expressing hemagglutinin (HA) of a H5N1 influenza virus in a dose escalation study in mice with the intranasal (IN) or intramuscular (IM) route of inoculation in comparison with the HAdV type C5 (HAdV-C5) vector (HAd-H5HA) expressing HA of a H5N1 influenza virus. Dose-related increases in the immune responses were clearly noticeable. A single IM inoculation with BAd-H5HA resulted in enhanced cellular immune responses compared with that of HAd-H5HA and conferred complete protection following challenge with a heterologous H5N1 virus at the dose of 3 × 107 plaque-forming units (PFUs), whereas a significant amount of influenza virus was detected in the lungs of mice immunized with 1 × 108 PFUs of HAd-H5HA. Similarly, compared with that of HAd-H5HA, a single IN inoculation with BAd-H5HA produced significantly enhanced humoral (HA-specific immunoglobulin [IgG] and its subclasses, as well as HA-specific IgA) and cellular immune responses, and conferred complete protection following challenge with a heterologous H5N1 virus. Complete protection with BAd-H5HA was observed with the lowest vaccine dose (1 × 106 PFUs), but similar protection with HAd-H5HA was observed at the highest vaccine dose (1 × 108 PFUs). These results suggest that at least 30-fold dose sparing can be achieved with BAd-H5HA vector compared with HAd-H5HA vaccine vector.
Collapse
|
15
|
Hassan AO, Vemula SV, Sharma A, Bangari DS, Mishra KK, Mittal SK. 155R is a novel structural protein of bovine adenovirus type 3, but it is not essential for virus replication. J Gen Virol 2017; 98:749-753. [PMID: 28086071 DOI: 10.1099/jgv.0.000707] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Bovine adenovirus (AdV) type 3 (BAdV-3) E1 region shares functional homology with E1 of human AdV type C5. Sequence analysis of the BAdV-3 E1 region revealed the presence of a novel 155R ORF that is not observed in other AdVs, on the lower strand antiparallel to a portion of the E1B region. The 155R gene products in BAdV-3-infected cells were identified by Northern blot, reverse transcriptase PCR followed by sequencing and Western blot analysis using the155R-specific antibody. 155R seems to be a late protein and is present in purified BAdV-3 particles. Replication kinetics of BAdV mutants with either one (BAdV/155R/mt1) or two (BAdV/155R/mt2) stop codons in the 155R ORF were comparable to those of BAdV-3, indicating that 155R is not essential for virus replication in cell culture. These results suggest that 155R-deleted BAdV-3 vectors could be generated in a cell line that fully complements BAdV-3 E1 functions.
Collapse
Affiliation(s)
- Ahmed O Hassan
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA.,Purdue Institute for Immunology, Inflammation and Infectious Diseases, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA.,Purdue University Center for Cancer Research, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
| | - Sai V Vemula
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA.,Purdue University Center for Cancer Research, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA.,Present address: Merck Sharp and Dohme, West Point, PA, USA
| | - Anurag Sharma
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA.,Purdue University Center for Cancer Research, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA.,Present address: Department of Pediatrics, Weill Cornell Medical College, New York, NY, USA
| | - Dinesh S Bangari
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA.,Purdue University Center for Cancer Research, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA.,Present address: Department of Pathology, Sanofi Genzyme, 5 Mountain Road, Framingham, MA, USA
| | - Krishna K Mishra
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA.,Purdue University Center for Cancer Research, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA.,Present address: Department of Biology, Ivy Tech Community College, Lafayette, IN, USA
| | - Suresh K Mittal
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA.,Purdue Institute for Immunology, Inflammation and Infectious Diseases, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA.,Purdue University Center for Cancer Research, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
| |
Collapse
|
16
|
Abstract
Antigenic drift of seasonal influenza viruses and the occasional introduction of influenza viruses of novel subtypes into the human population complicate the timely production of effective vaccines that antigenically match the virus strains that cause epidemic or pandemic outbreaks. The development of game-changing vaccines that induce broadly protective immunity against a wide variety of influenza viruses is an unmet need, in which recombinant viral vectors may provide. Use of viral vectors allows the delivery of any influenza virus antigen, or derivative thereof, to the immune system, resulting in the optimal induction of virus-specific B- and T-cell responses against this antigen of choice. This systematic review discusses results obtained with vectored influenza virus vaccines and advantages and disadvantages of the currently available viral vectors.
Collapse
Affiliation(s)
- Rory D de Vries
- a Department of Viroscience , Erasmus MC , Rotterdam , The Netherlands
| | | |
Collapse
|
17
|
Xiang K, Ying G, Yan Z, Shanshan Y, Lei Z, Hongjun L, Maosheng S. Progress on adenovirus-vectored universal influenza vaccines. Hum Vaccin Immunother 2016; 11:1209-22. [PMID: 25876176 DOI: 10.1080/21645515.2015.1016674] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Influenza virus (IFV) infection causes serious health problems and heavy financial burdens each year worldwide. The classical inactivated influenza virus vaccine (IIVV) and live attenuated influenza vaccine (LAIV) must be updated regularly to match the new strains that evolve due to antigenic drift and antigenic shift. However, with the discovery of broadly neutralizing antibodies that recognize conserved antigens, and the CD8(+) T cell responses targeting viral internal proteins nucleoprotein (NP), matrix protein 1 (M1) and polymerase basic 1 (PB1), it is possible to develop a universal influenza vaccine based on the conserved hemagglutinin (HA) stem, NP, and matrix proteins. Recombinant adenovirus (rAd) is an ideal influenza vaccine vector because it has an ideal stability and safety profile, induces balanced humoral and cell-mediated immune responses due to activation of innate immunity, provides 'self-adjuvanting' activity, can mimic natural IFV infection, and confers seamless protection against mucosal pathogens. Moreover, this vector can be developed as a low-cost, rapid-response vaccine that can be quickly manufactured. Therefore, an adenovirus vector encoding conserved influenza antigens holds promise in the development of a universal influenza vaccine. This review will summarize the progress in adenovirus-vectored universal flu vaccines and discuss future novel approaches.
Collapse
Key Words
- ADCC, antibody-dependent cell-mediated cytotoxicity
- APC, antigen-presenting cell
- Ad: adenovirus
- CAR, Coxsackie-Adenovirus Receptor
- CTLs, cytotoxic T lymphocytes
- DC, lung dendritic cells
- DVD, drug–vaccine duo
- FcγRs, Fc receptors for IgG
- HA, hemagglutinin
- HDAd, helper-dependent adenoviral
- HEK293, human embryonic kidney 293 cell
- HI, hemagglutination inhibition
- HLA, human leukocyte antigen
- IF-γ, interferon-γ
- IFV, Influenza virus
- IIVV, inactivated influenza virus vaccine
- IL-2, interleukin-2
- ITRs, inverted terminal repeats
- LAIV, live attenuated influenza vaccine
- M1, matrix protein 1
- M2, matrix protein 2
- MHC-I, major histocompatibility complex class I
- NA, neuraminidase
- NP, nucleoprotein
- RCA, replication competent adenovirus
- VAERD, vaccine-associated enhanced respiratory disease
- adenovirus vector
- broadly neutralizing antibodies
- cellular immunity
- flu, influenza
- hemagglutinin
- humoral immunity
- influenza
- mAbs, monoclonal antibodies
- mucosal immunity
- rAd, recombinant adenovirus
- universal vaccine
Collapse
Affiliation(s)
- Kui Xiang
- a Department of Molecular Biology; Institute of Medical Biology; Chinese Academy of Medical Sciences; Peking Union Medical College ; Kunming , Yunnan , PR China
| | | | | | | | | | | | | |
Collapse
|
18
|
A highly immunogenic vaccine against A/H7N9 influenza virus. Vaccine 2016; 34:744-9. [PMID: 26765287 DOI: 10.1016/j.vaccine.2015.12.062] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 12/09/2015] [Accepted: 12/24/2015] [Indexed: 11/23/2022]
Abstract
Since the first case of human infection in March 2013, continued reports of H7N9 cases highlight a potential pandemic threat. Highly immunogenic vaccines to this virus are urgently needed to protect vulnerable populations who lack protective immunity. In this study, an egg- and adjuvant-independent adenoviral vector-based, hemagglutinin H7 subtype influenza vaccine (HAd-H7HA) demonstrated enhanced cell-mediated immunity as well as serum antibody responses in a mouse model. Most importantly, this vaccine provided complete protection against homologous A/H7N9 viral challenge suggesting its potential utility as a pandemic vaccine.
Collapse
|
19
|
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
|
20
|
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
|
21
|
Bitrus Y, Andrew JN, Owolodun OA, Luka PD, Umaru DA. The reoccurrence of H5N1 outbreaks necessitates the development of safe and effective influenza vaccine technologies for the prevention and control of avian influenza in Sub-Saharan Africa. ACTA ACUST UNITED AC 2015. [DOI: 10.5897/bmbr2015.0246] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
22
|
He B, Zheng BJ, Wang Q, Du L, Jiang S, Lu L. Adenovirus-based vaccines against avian-origin H5N1 influenza viruses. Microbes Infect 2015; 17:135-41. [PMID: 25479556 PMCID: PMC7110517 DOI: 10.1016/j.micinf.2014.11.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 11/18/2014] [Accepted: 11/18/2014] [Indexed: 02/03/2023]
Abstract
Since 1997, human infection with avian H5N1, having about 60% mortality, has posed a threat to public health. In this review, we describe the epidemiology of H5N1 transmission, advantages and disadvantages of different influenza vaccine types, and characteristics of adenovirus, finally summarizing advances in adenovirus-based H5N1 systemic and mucosal vaccines.
Collapse
Affiliation(s)
- Biao He
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Shanghai Medical College and Institute of Medical Microbiology, Fudan University, Shanghai 200032, China
| | - Bo-jian Zheng
- Department of Microbiology, University of Hong Kong, Pokfulam, Hong Kong 999077, China
| | - Qian Wang
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Shanghai Medical College and Institute of Medical Microbiology, Fudan University, Shanghai 200032, China
| | - Lanying Du
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY 10065, USA
| | - Shibo Jiang
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Shanghai Medical College and Institute of Medical Microbiology, Fudan University, Shanghai 200032, China; Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY 10065, USA.
| | - Lu Lu
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Shanghai Medical College and Institute of Medical Microbiology, Fudan University, Shanghai 200032, China.
| |
Collapse
|
23
|
Majhen D, Calderon H, Chandra N, Fajardo CA, Rajan A, Alemany R, Custers J. Adenovirus-based vaccines for fighting infectious diseases and cancer: progress in the field. Hum Gene Ther 2014; 25:301-17. [PMID: 24580050 DOI: 10.1089/hum.2013.235] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
The field of adenovirology is undergoing rapid change in response to increasing appreciation of the potential advantages of adenoviruses as the basis for new vaccines and as vectors for gene and cancer therapy. Substantial knowledge and understanding of adenoviruses at a molecular level has made their manipulation for use as vaccines and therapeutics relatively straightforward in comparison with other viral vectors. In this review we summarize the structure and life cycle of the adenovirus and focus on the use of adenovirus-based vectors in vaccines against infectious diseases and cancers. Strategies to overcome the problem of preexisting antiadenovirus immunity, which can hamper the immunogenicity of adenovirus-based vaccines, are discussed. When armed with tumor-associated antigens, replication-deficient and oncolytic adenoviruses can efficiently activate an antitumor immune response. We present concepts on how to use adenoviruses as therapeutic cancer vaccines and consider some of the strategies used to further improve antitumor immune responses. Studies that explore the prospect of adenoviruses as vaccines against infectious diseases and cancer are underway, and here we give an overview of the latest developments.
Collapse
|
24
|
Cheng L, Huang X, Li X, Xiong W, Sun W, Yang C, Zhang K, Wang Y, Liu H, Huang X, Ji G, Sun F, Zheng C, Zhu P. Cryo-EM structures of two bovine adenovirus type 3 intermediates. Virology 2014; 450-451:174-81. [PMID: 24503080 DOI: 10.1016/j.virol.2013.12.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Revised: 10/04/2013] [Accepted: 12/10/2013] [Indexed: 01/28/2023]
Abstract
Adenoviruses (Ads) infect hosts from all vertebrate species and have been investigated as vaccine vectors. We report here near-atomic structures of two bovine Ad type 3 (BAd3) intermediates obtained by cryo-electron microscopy. A comparison between the two intermediate structures reveals that the differences are localized in the fivefold vertex region, while their facet structures are identical. The overall facet structure of BAd3 exhibits a similar structure to human Ads; however, BAd3 protein IX has a unique conformation. Mass spectrometry and cryo-electron tomography analyses indicate that one intermediate structure represents the stage during DNA encapsidation, whilst the other intermediate structure represents a later stage. These results also suggest that cleavage of precursor protein VI occurs during, rather than after, the DNA encapsidation process. Overall, our results provide insights into the mechanism of Ad assembly, and allow the first structural comparison between human and nonhuman Ads at backbone level.
Collapse
Affiliation(s)
- Lingpeng Cheng
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Xiaoxing Huang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Xiaomin Li
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Wei Xiong
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Luo-jia-shan, Wuhan, Hubei 430072, China
| | - Wei Sun
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Chongwen Yang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Kai Zhang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Ying Wang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Hongrong Liu
- College of Physics and Information Science, Hunan Normal University, Changsha, Hunan 410081, China
| | - Xiaojun Huang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Gang Ji
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Fei Sun
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Congyi Zheng
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Luo-jia-shan, Wuhan, Hubei 430072, China.
| | - Ping Zhu
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.
| |
Collapse
|
25
|
Lukashevich IS, Shirwan H. Adenovirus-Based Vectors for the Development of Prophylactic and Therapeutic Vaccines. NOVEL TECHNOLOGIES FOR VACCINE DEVELOPMENT 2014. [PMCID: PMC7121347 DOI: 10.1007/978-3-7091-1818-4_8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Emerging and reemerging infectious diseases as well as cancer pose great global health impacts on the society. Vaccines have emerged as effective treatments to prevent or reduce the burdens of already developed diseases. This is achieved by means of activating various components of the immune system to generate systemic inflammatory reactions targeting infectious agents or diseased cells for control/elimination. DNA virus-based genetic vaccines gained significant attention in the past decades owing to the development of DNA manipulation technologies, which allowed engineering of recombinant viral vectors encoding sequences for foreign antigens or their immunogenic epitopes as well as various immunomodulatory molecules. Despite tremendous progress in the past 50 years, many hurdles still remain for achieving the full clinical potential of viral-vectored vaccines. This chapter will present the evolution of vaccines from “live” or “attenuated” first-generation agents to recombinant DNA and viral-vectored vaccines. Particular emphasis will be given to human adenovirus (Ad) for the development of prophylactic and therapeutic vaccines. Ad biological properties related to vaccine development will be highlighted along with their advantages and potential hurdles to be overcome. In particular, we will discuss (1) genetic modifications in the Ad capsid protein to reduce the intrinsic viral immunogenicity, (2) antigen capsid incorporation for effective presentation of foreign antigens to the immune system, (3) modification of the hexon and fiber capsid proteins for Ad liver de-targeting and selective retargeting to cancer cells, (4) Ad-based vaccines carrying “arming” transgenes with immunostimulatory functions as immune adjuvants, and (5) oncolytic Ad vectors as a new therapeutic approach against cancer. Finally, the combination of adenoviral vectors with other non-adenoviral vector systems, the prime/boost strategy of immunization, clinical trials involving Ad-based vaccines, and the perspectives for the field development will be discussed.
Collapse
Affiliation(s)
- Igor S Lukashevich
- Department of Pharmacology and Toxicolog Department of Microbiology and Immunolog, University of Louisville, Louisville, Kentucky USA
| | - Haval Shirwan
- Department of Microbiology and Immunolog, University of Louisville, Louisville, Kentucky USA
| |
Collapse
|
26
|
Abstract
Ebola is a highly virulent pathogen causing severe hemorrhagic fever with a high case fatality rate in humans and non-human primates (NHPs). Although safe and effective vaccines or other medicinal agents to block Ebola infection are currently unavailable, a significant effort has been put forth to identify several promising candidates for the treatment and prevention of Ebola hemorrhagic fever. Among these, recombinant adenovirus-based vectors have been identified as potent vaccine candidates, with some affording both pre- and post-exposure protection from the virus. Recently, Investigational New Drug (IND) applications have been approved by the US Food and Drug Administration (FDA) and phase I clinical trials have been initiated for two small-molecule therapeutics: anti-sense phosphorodiamidate morpholino oligomers (PMOs: AVI-6002, AVI-6003) and lipid nanoparticle/small interfering RNA (LNP/siRNA: TKM-Ebola). These potential alternatives to vector-based vaccines require multiple doses to achieve therapeutic efficacy, which is not ideal with regard to patient compliance and outbreak scenarios. These concerns have fueled a quest for even better vaccination and treatment strategies. Here, we summarize recent advances in vaccines or post-exposure therapeutics for prevention of Ebola hemorrhagic fever. The utility of novel pharmaceutical approaches to refine and overcome barriers associated with the most promising therapeutic platforms are also discussed.
Collapse
Affiliation(s)
- Jin Huk Choi
- Division of Pharmaceutics, College of Pharmacy, The University of Texas at Austin, Austin, TX, 78712, U.S.A
| | - Maria A. Croyle
- Division of Pharmaceutics, College of Pharmacy, The University of Texas at Austin, Austin, TX, 78712, U.S.A
- Institute of Cellular and Molecular Biology, The University of Texas at Austin, Austin, TX, 78712, U.S.A
| |
Collapse
|
27
|
Vemula SV, Amen O, Katz JM, Donis R, Sambhara S, Mittal SK. Beta-defensin 2 enhances immunogenicity and protection of an adenovirus-based H5N1 influenza vaccine at an early time. Virus Res 2013; 178:398-403. [PMID: 24051000 DOI: 10.1016/j.virusres.2013.09.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Revised: 08/30/2013] [Accepted: 09/06/2013] [Indexed: 11/16/2022]
Abstract
Reports of human infections with highly pathogenic H5N1 avian influenza viruses in many countries in Asia and Africa with varying case fatality rates highlight the pandemic potential of these viruses. In order to contain a rapidly spreading influenza virus in a pandemic scenario, a vaccine which can induce rapid and robust immune responses, preferably in a single dose, is necessary. Murine beta-defensin 2 (Mbd2), a small molecular weight protein expressed by epithelial cells, has been shown to enhance antigen-specific immune responses by recruiting and activating professional antigen presenting cells to the site of vaccination. This study assessed the potential of Mbd2 to enhance the immunogenicity and protective efficacy of a human adenovirus (HAd)-based vaccine expressing the hemagglutinin (HA) and nucleoprotein (NP) [HAd-HA-NP] of an H5N1 influenza virus. A single inoculation of mice with both HAd-HA-NP and a HAd vector expressing Murine β-defensin 2 (HAd-Mbd2) resulted in significantly higher levels of both humoral and cell-mediated immune responses compared to the groups vaccinated only with HAd-HA-NP. These responses were evident even at day 7 post-immunization. Furthermore, the HAd-HA-NP+HAd-Mbd2-immunized group receiving the lowest vector dose (2 × 10(7)+1 × 10(7)) was completely protected against an rgH5N1 virus challenge on day 7 post-vaccination. These results highlight the potential of Mbd2 as a genetic adjuvant in inducing rapid and robust immune responses to a HAd-based vaccine.
Collapse
Affiliation(s)
- Sai V Vemula
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA; Bindley Bioscience Center, Purdue University, West Lafayette, IN, USA
| | | | | | | | | | | |
Collapse
|
28
|
Vemula SV, Ahi YS, Swaim AM, Katz JM, Donis R, Sambhara S, Mittal SK. Broadly protective adenovirus-based multivalent vaccines against highly pathogenic avian influenza viruses for pandemic preparedness. PLoS One 2013; 8:e62496. [PMID: 23638099 PMCID: PMC3640067 DOI: 10.1371/journal.pone.0062496] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Accepted: 03/22/2013] [Indexed: 11/19/2022] Open
Abstract
Recurrent outbreaks of H5, H7 and H9 avian influenza viruses in domestic poultry accompanied by their occasional transmission to humans have highlighted the public health threat posed by these viruses. Newer vaccine approaches for pandemic preparedness against these viruses are needed, given the limitations of vaccines currently approved for H5N1 viruses in terms of their production timelines and the ability to induce protective immune responses in the absence of adjuvants. In this study, we evaluated the feasibility of an adenovirus (AdV)-based multivalent vaccine approach for pandemic preparedness against H5, H7 and H9 avian influenza viruses in a mouse model. Replication-defective AdV vectors expressing hemagglutinin (HA) from different subtypes and nucleoprotein (NP) from one subtype induced high levels of humoral and cellular immune responses and conferred protection against virus replication following challenge with H5, H7 and H9 avian influenza virus subtypes. Inclusion of HA from the 2009 H1N1 pandemic virus in the vaccine formulation further broadened the vaccine coverage. Significantly high levels of HA stalk-specific antibodies were observed following immunization with the multivalent vaccine. Inclusion of NP into the multivalent HA vaccine formulation resulted in the induction of CD8 T cell responses. These results suggest that a multivalent vaccine strategy may provide reasonable protection in the event of a pandemic caused by H5, H7, or H9 avian influenza virus before a strain-matched vaccine can be produced.
Collapse
Affiliation(s)
- Sai V. Vemula
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana, United States of America
- Bindley Bioscience Center, Purdue University, West Lafayette, Indiana, United States of America
| | - Yadvinder S. Ahi
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana, United States of America
- Bindley Bioscience Center, Purdue University, West Lafayette, Indiana, United States of America
| | - Anne-Marie Swaim
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana, United States of America
- Bindley Bioscience Center, Purdue University, West Lafayette, Indiana, United States of America
| | - Jacqueline M. Katz
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Ruben Donis
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Suryaprakash Sambhara
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
- * E-mail: (SM); (SS)
| | - Suresh K. Mittal
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana, United States of America
- Bindley Bioscience Center, Purdue University, West Lafayette, Indiana, United States of America
- * E-mail: (SM); (SS)
| |
Collapse
|
29
|
Airway delivery of an adenovirus-based Ebola virus vaccine bypasses existing immunity to homologous adenovirus in nonhuman primates. J Virol 2013; 87:3668-77. [PMID: 23302894 DOI: 10.1128/jvi.02864-12] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Anti-adenovirus serotype 5 antibodies are capable of neutralizing adenovirus serotype 5-based vaccines. In mice and guinea pigs, intranasal delivery of adenovirus serotype 5-based vaccine bypasses induced adenovirus serotype 5 preexisting immunity, resulting in protection against species-adapted Ebola virus challenge. In this study, nonhuman primates were vaccinated with adenovirus serotype 5-based vaccine either intramuscularly or via the airway route (intranasally/intratracheally) in the presence or absence of adenovirus serotype 5 preexisting immunity. Immune responses were evaluated to determine the effect of both the vaccine delivery route and preexisting immunity before and after a lethal Ebola virus (Zaïre strain Kikwit 95) challenge. Intramuscular vaccination fully protected nonhuman primates in the absence of preexisting immunity, whereas the presence of preexisting immunity abrogated vaccine efficacy and resulted in complete mortality. In contrast, the presence of preexisting immunity to adenovirus serotype 5 did not alter the survival rate of nonhuman primates receiving the adenovirus serotype 5-based Ebola virus vaccine in the airway. This study shows that airway vaccination with adenovirus serotype 5-based Ebola virus vaccine can efficiently bypass preexisting immunity to adenovirus serotype 5 and induce protective immune responses, albeit at lower efficacy than that using an intramuscular vaccine delivery route.
Collapse
|
30
|
Mooney AJ, Tompkins SM. Experimental vaccines against potentially pandemic and highly pathogenic avian influenza viruses. Future Virol 2013; 8:25-41. [PMID: 23440999 PMCID: PMC3579652 DOI: 10.2217/fvl.12.122] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Influenza A viruses continue to emerge and re-emerge, causing outbreaks, epidemics and occasionally pandemics. While the influenza vaccines licensed for public use are generally effective against seasonal influenza, issues arise with production, immunogenicity, and efficacy in the case of vaccines against pandemic and emerging influenza viruses, and highly pathogenic avian influenza virus in particular. Thus, there is need of improved influenza vaccines and vaccination strategies. This review discusses advances in alternative influenza vaccines, touching briefly on licensed vaccines and vaccine antigens; then reviewing recombinant subunit vaccines, virus-like particle vaccines and DNA vaccines, with the main focus on virus-vectored vaccine approaches.
Collapse
Affiliation(s)
- Alaina J Mooney
- Department of Infectious Diseases, University of Georgia, 111 Carlton St, Athens, GA 30602, USA
| | - S Mark Tompkins
- Department of Infectious Diseases, University of Georgia, 111 Carlton St, Athens, GA 30602, USA
| |
Collapse
|
31
|
Meng Q, Lin Y, Ma J, Ma Y, Zhao L, Li S, Yang K, Zhou J, Shen R, Zhang X, Shao Y. A pilot study comparing the development of EIAV Env-specific antibodies induced by DNA/recombinant vaccinia-vectored vaccines and an attenuated Chinese EIAV vaccine. Viral Immunol 2012; 25:477-84. [PMID: 23171359 DOI: 10.1089/vim.2012.0014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Data from successful attenuated lentiviral vaccine studies indicate that fully mature Env-specific antibodies characterized by high titer, high avidity, and the predominant recognition of conformational epitopes are associated with protective efficacy. Although vaccination with a DNA prime/recombinant vaccinia-vectored vaccine boost strategy has been found to be effective in some trials with non-human primate/simian/human immunodeficiency virus (SHIV) models, it remains unclear whether this vaccination strategy could elicit mature equine infectious anemia virus (EIAV) Env-specific antibodies, thus protecting vaccinated horses against EIAV infection. Therefore, in this pilot study we vaccinated horses using a strategy based on DNA prime/recombinant Tiantan vaccinia (rTTV)-vectored vaccines encoding EIAV env and gag genes, and observed the development of Env-specific antibodies, neutralizing antibodies, and p26-specific antibodies. Vaccination with DNA induced low titer, low avidity, and the predominant recognition of linear epitopes by Env-specific antibodies, which was enhanced by boosting vaccinations with rTTV vaccines. However, the maturation levels of Env-specific antibodies induced by the DNA/rTTV vaccines were significantly lower than those induced by the attenuated vaccine EIAV(FDDV). Additionally, DNA/rTTV vaccines did not elicit broadly neutralizing antibodies. After challenge with a virulent EIAV strain, all of the vaccinees and control horses died from EIAV disease. These data indicate that the regimen of DNA prime/rTTV vaccine boost did not induce mature Env-specific antibodies, which might have contributed to immune protection failure.
Collapse
Affiliation(s)
- Qinglai Meng
- State Key Laboratory for Infectious Disease Prevention and Control, National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Patel A, Kobinger GP. Evaluation of mismatched immunity against influenza viruses. Future Virol 2012. [DOI: 10.2217/fvl.12.105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Prior immunity against influenza A viruses generates sterilizing immunity against matched (homologous) viruses and varying levels of protection against mismatched (heterologous) viruses of the same or different subtypes. Natural immunity carries the risk of high morbidity and mortality, therefore immunization offers the best preventative measure. Antibody responses against the viral hemagglutinin protein correlate with protection in humans and evidence increasingly supports a role for robust cellular immune responses. By exploiting mismatched immunity, current conventional and experimental vaccine candidates can improve the generation of cross-protective immune responses against heterologous viruses. Experimental vaccines such as virus-like particles, DNA vectors, viral vectors and broadly neutralizing antibodies are able to expand cross-protection through mismatched B- and T-cell responses. However, the generation of mismatched immune responses can also have the opposite effect and impair protective immunity. This review discusses mismatched immunity in the context of natural infection and immunization. Additionally, we discuss strategies to exploit mismatched immunity in order to improve current conventional and experimental influenza A virus vaccines.
Collapse
Affiliation(s)
- Ami Patel
- San Raffaele-Telethon Institute of Gene Therapy (hSR-TIGET), Milan, Italy Division of Gene Therapy & Regenerative Medicine, via Olgettina 58, Milan, Italy, 20132
| | - Gary P Kobinger
- Department of Medical Microbiology, University of Manitoba, Winnipeg, Manitoba, Canada
- Department of Immunology, University of Manitoba, Winnipeg, Manitoba, Canada
- Special Pathogens Programme, National Microbiology Laboratory, Public Health Agency of Canada, Canadian Science Centre for Human & Animal Health, 1015 Arlington Street, Winnipeg, Manitoba, Canada, R3E 3R2
| |
Collapse
|
33
|
Abstract
The emergence of a highly pathogenic avian influenza virus H5N1 has increased the potential for a new pandemic to occur. This event highlights the necessity for developing a new generation of influenza vaccines to counteract influenza disease. These vaccines must be manufactured for mass immunization of humans in a timely manner. Poultry should be included in this policy, since persistent infected flocks are the major source of avian influenza for human infections. Recombinant adenoviral vectored H5N1 vaccines are an attractive alternative to the currently licensed influenza vaccines. This class of vaccines induces a broadly protective immunity against antigenically distinct H5N1, can be manufactured rapidly, and may allow mass immunization of human and poultry. Recombinant adenoviral vectors derived from both human and non-human adenoviruses are currently being investigated and appear promising both in nonclinical and clinical studies. This review will highlight the current status of various adenoviral vectored H5N1 vaccines and will outline novel approaches for the future.
Collapse
|
34
|
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
|
35
|
Pandey A, Singh N, Vemula SV, Couëtil L, Katz JM, Donis R, Sambhara S, Mittal SK. Impact of preexisting adenovirus vector immunity on immunogenicity and protection conferred with an adenovirus-based H5N1 influenza vaccine. PLoS One 2012; 7:e33428. [PMID: 22432020 PMCID: PMC3303828 DOI: 10.1371/journal.pone.0033428] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Accepted: 02/10/2012] [Indexed: 12/20/2022] Open
Abstract
The prevalence of preexisting immunity to adenoviruses in the majority of the human population might adversely impact the development of adaptive immune responses against adenovirus vector-based vaccines. To address this issue, we primed BALB/c mice either intranasally (i.n.) or intramuscularly (i.m.) with varying doses of wild type (WT) human adenovirus subtype 5 (HAd5). Following the development of immunity against HAd5, we immunized animals via the i.n. or i.m. route of inoculation with a HAd vector (HAd-HA-NP) expressing the hemagglutinin (HA) and nucleoprotein (NP) of A/Vietnam/1203/04 (H5N1) influenza virus. The immunogenicity and protection results suggest that low levels of vector immunity (<520 virus-neutralization titer) induced by priming mice with up to 10(7) plaque forming units (p.f.u.) of HAd-WT did not adversely impact the protective efficacy of the vaccine. Furthermore, high levels of vector immunity (approximately 1500 virus-neutralization titer) induced by priming mice with 10(8) p.f.u. of HAd-WT were overcome by either increasing the vaccine dose or using alternate routes of vaccination. A further increase in the priming dose to 10(9) p.f.u. allowed only partial protection. These results suggest possible strategies to overcome the variable levels of human immunity against adenoviruses, leading to better utilization of HAd vector-based vaccines.
Collapse
Affiliation(s)
- Aseem Pandey
- Department of Comparative Pathobiology, Purdue University, West Lafayette, Indiana, United States of America
- Bindley Bioscience Center, Purdue University, West Lafayette, Indiana, United States of America
| | - Neetu Singh
- Department of Comparative Pathobiology, Purdue University, West Lafayette, Indiana, United States of America
- Bindley Bioscience Center, Purdue University, West Lafayette, Indiana, United States of America
| | - Sai V. Vemula
- Department of Comparative Pathobiology, Purdue University, West Lafayette, Indiana, United States of America
- Bindley Bioscience Center, Purdue University, West Lafayette, Indiana, United States of America
| | - Laurent Couëtil
- Department of Clinical Veterinary Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana, United States of America
| | - Jacqueline M. Katz
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Ruben Donis
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Suryaprakash Sambhara
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
- * E-mail: (SS); (SKM)
| | - Suresh K. Mittal
- Department of Comparative Pathobiology, Purdue University, West Lafayette, Indiana, United States of America
- Bindley Bioscience Center, Purdue University, West Lafayette, Indiana, United States of America
- * E-mail: (SS); (SKM)
| |
Collapse
|
36
|
Richardson JS, Abou MC, Tran KN, Kumar A, Sahai BM, Kobinger GP. Impact of systemic or mucosal immunity to adenovirus on Ad-based Ebola virus vaccine efficacy in guinea pigs. J Infect Dis 2011; 204 Suppl 3:S1032-42. [PMID: 21987739 DOI: 10.1093/infdis/jir332] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Approximately 35% of the North American population and an estimated 90% of the sub-Saharan African population have antibodies against adenovirus serotype 5 (AdHu5) that are capable of neutralizing AdHu5-based vaccines. In mice, intranasal delivery of AdHu5 expressing the Zaire ebolavirus glycoprotein human adenovirus serotype 5 (Ad) containing the genes for the Zaire ebolavirus glycoprotein (ZGP) under the expressional control of a cytomegalovirus immediate early promoter (CMV)) can bypass systemic preexisting immunity, resulting in protection against mouse-adapted Zaire ebolavirus (Mayinga 1976). METHODS Guinea pigs administered an adenovirus-based Ebola virus vaccine either intramuscularly or intranasally in the presence of systemically or mucosally induced adenovirus immunity were challenged with a lethal dose of guinea pig-adapted Zaire ebolavirus (Mayinga 1976) (GA-ZEBOV). The humoral immune response was assayed to determine the effect of vaccine delivery route and preexisting immunity. RESULTS Intramuscular or intranasal vaccination fully protected guinea pigs against a lethal GA-ZEBOV challenge. However, intramuscular vaccination in animals with systemically induced preexisting immunity resulted in low survival following challenge. Interestingly, intranasal vaccination protected guinea pigs with systemic preexisting immunity to AdHu5. Mucosal adenoviral immunity induced by intranasal administration of AdHu5 decreased protection following intranasal vaccination with the first-generation but not with the second-generation vaccine. CONCLUSIONS Intranasal vaccination is an effective vaccine delivery route in the presence of systemic and, to a lower extent, mucosal preexisting immunity to the vaccine vector in guinea pigs.
Collapse
Affiliation(s)
- Jason S Richardson
- Special Pathogens Department, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada
| | | | | | | | | | | |
Collapse
|
37
|
Sequential administration of bovine and human adenovirus vectors to overcome vector immunity in an immunocompetent mouse model of breast cancer. Virus Res 2011; 163:202-11. [PMID: 21971215 DOI: 10.1016/j.virusres.2011.09.031] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2011] [Revised: 09/20/2011] [Accepted: 09/20/2011] [Indexed: 11/24/2022]
Abstract
The potential of a bovine adenovirus serotype 3 (BAd3)-based vector to bypass the human adenoviral serotype 5 (HAd5)-specific neutralizing immune response was evaluated in an immunocompetent mouse model of breast cancer. Initially we monitored vector biodistribution, genome persistence, transgene expression, and potential toxicity of HAd-GFP [HAd5 vector expressing green fluorescent protein (GFP)] or BAd-GFP (BAd3 vector expressing GFP) in FVB/n mice bearing tumors. A comparable biodistribution pattern for BAd-GFP and HAd-GFP was evident. In addition, following the development of vector-specific immune responses, animals were inoculated intratumorally (i.t.) with HAd-GFP or BAd-GFP. HAd-GFP immunity did not hamper the transduction and persistence of BAd-GFP into the tumors and other organs, and, similarly, BAd-GFP immunity did not hamper the transduction and persistence of HAd-GFP. Both BAd3 and HAd5 vectors showed relatively higher transgene expression in the presence of heterologous vector immunity. In contrast, the homologous vector immunity was associated with a rapid vector clearance and decline in transgene expression levels. Histopathological changes in BAd-GFP inoculated animals were generally mild with some acute but recoverable hepatic perturbations. Overall, the data suggest the importance of BAd3 vectors for sequential vector administration in overcoming the vector immunity for cancer gene therapy.
Collapse
|
38
|
Vemula SV, Mittal SK. Production of adenovirus vectors and their use as a delivery system for influenza vaccines. Expert Opin Biol Ther 2011; 10:1469-87. [PMID: 20822477 DOI: 10.1517/14712598.2010.519332] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
IMPORTANCE OF THE FIELD With the emergence of highly pathogenic avian influenza H5N1 viruses that have crossed species barriers and are responsible for lethal infections in humans in many countries, there is an urgent need for the development of effective vaccines which can be produced in large quantities at a short notice and confer broad protection against these H5N1 variants. In order to meet the potential global vaccine demand in a pandemic scenario, new vaccine-production strategies must be explored in addition to the currently used egg-based technology for seasonal influenza. AREAS COVERED IN THIS REVIEW Adenovirus (Ad) based influenza vaccines represent an attractive alternative/supplement to the currently licensed egg-based influenza vaccines. Ad-based vaccines are relatively inexpensive to manufacture, and their production process does not require either chicken eggs or labor-intensive and time-consuming processes necessitating enhanced biosafety facilities. Most importantly, in a pandemic situation, this vaccine strategy could offer a stockpiling option to reduce the response time before a strain-matched vaccine could be developed. WHAT THE READER WILL GAIN This review discusses Ad-vector technology and the current progress in the development of Ad-based influenza vaccines. TAKE HOME MESSAGE Ad vector-based influenza vaccines for pandemic preparedness are under development to meet global vaccine demand.
Collapse
Affiliation(s)
- Sai V Vemula
- Purdue University, Bindley Bioscience Center, School of Veterinary Medicine, Department of Comparative Pathobiology, West Lafayette, IN 47907, USA
| | | |
Collapse
|
39
|
Lanzi A, Ben Youssef G, Perricaudet M, Benihoud K. Anti-adenovirus humoral responses influence on the efficacy of vaccines based on epitope display on adenovirus capsid. Vaccine 2010; 29:1463-71. [PMID: 21184856 DOI: 10.1016/j.vaccine.2010.12.025] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2010] [Revised: 11/26/2010] [Accepted: 12/09/2010] [Indexed: 11/30/2022]
Abstract
The efficacy of recombinant adenoviruses (Ads) vaccine vectors is diminished by the high prevalence of anti-Ad antibodies (Abs) that hampers gene transfer. Epitope display on Ad capsid constitutes an alternative approach to bypass anti-Ad Ab capacity from blocking antigen expression. To understand the role of the epitope insertion site, an ovalbumin-derived epitope was genetically inserted into either Ad hexon or fiber proteins. Hexon-modified Ads triggered higher anti-ovalbumin Ab responses after one injection but surprisingly fiber-modified Ads were by far more potent after two or several administrations. Our data unravel a role for anti-Ad humoral immunity in controlling anti-epitope humoral responses.
Collapse
Affiliation(s)
- Anastasia Lanzi
- CNRS UMR 8203, Vectorologie et thérapeutiques anticancéreuses, Institut Gustave Roussy, 39 rue Camille Desmoulins, 94805 Villejuif Cedex, France
| | | | | | | |
Collapse
|
40
|
Patel A, Tikoo S, Kobinger G. A porcine adenovirus with low human seroprevalence is a promising alternative vaccine vector to human adenovirus 5 in an H5N1 virus disease model. PLoS One 2010; 5:e15301. [PMID: 21179494 PMCID: PMC3002947 DOI: 10.1371/journal.pone.0015301] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2010] [Accepted: 11/10/2010] [Indexed: 12/22/2022] Open
Abstract
Human adenovirus 5 (AdHu5) vectors are robust vaccine platforms however the presence of naturally-acquired neutralizing antibodies may reduce vector efficacy and potential for re-administration. This study evaluates immune responses and protection following vaccination with a replication-incompetent porcine adenovirus 3 (PAV3) vector as an alternative vaccine to AdHu5 using an avian influenza H5N1 disease model. Vaccine efficacy was evaluated in BALB/c mice following vaccination with different doses of the PAV3 vector expressing an optimized A/Hanoi/30408/2005 H5N1 hemagglutinin antigen (PAV3-HA) and compared with an AdHu5-HA control. PAV3-HA rapidly generated antibody responses, with significant neutralizing antibody titers on day 21, and stronger cellular immune responses detected on day 8, compared to AdHu5-HA. The PAV3-HA vaccine, administered 8 days before challenge, demonstrated improved survival and lower virus load. Evaluation of long-term vaccine efficacy at 12 months post-vaccination showed better protection with the PAV3-HA than with the AdHu5-HA vaccine. Importantly, as opposed to AdHu5, PAV3 vector was not significantly neutralized by human antibodies pooled from over 10,000 individuals. Overall, PAV3-based vector is capable of mediating swift, strong immune responses and offer a promising alternative to AdHu5.
Collapse
Affiliation(s)
- Ami Patel
- National Microbiology Laboratory, Public Health Agency of Canada, Canadian Science Centre for Human and Animal Health, Winnipeg, Canada
- Department of Medical Microbiology, University of Manitoba, Winnipeg, Canada
| | - Suresh Tikoo
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, Canada
| | - Gary Kobinger
- National Microbiology Laboratory, Public Health Agency of Canada, Canadian Science Centre for Human and Animal Health, Winnipeg, Canada
- Department of Medical Microbiology, University of Manitoba, Winnipeg, Canada
- * E-mail:
| |
Collapse
|
41
|
Chen H, Xiang ZQ, Li Y, Kurupati RK, Jia B, Bian A, Zhou DM, Hutnick N, Yuan S, Gray C, Serwanga J, Auma B, Kaleebu P, Zhou X, Betts MR, Ertl HCJ. Adenovirus-based vaccines: comparison of vectors from three species of adenoviridae. J Virol 2010; 84:10522-32. [PMID: 20686035 PMCID: PMC2950567 DOI: 10.1128/jvi.00450-10] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2010] [Accepted: 07/27/2010] [Indexed: 11/20/2022] Open
Abstract
In order to better understand the broad applicability of adenovirus (Ad) as a vector for human vaccine studies, we compared four adenovirus (Ad) vectors from families C (Ad human serotype 5 [HAdV-5; here referred to as AdHu5]), D (HAdV-26; here referred to as AdHu26), and E (simian serotypes SAdV-23 and SAdV-24; here referred to as chimpanzee serotypes 6 and 7 [AdC6 and AdC7, respectively]) of the Adenoviridae. Seroprevalence rates and titers of neutralizing antibodies to the two human-origin Ads were found to be higher than those reported previously, especially in countries of sub-Saharan Africa. Conversely, prevalence rates and titers to AdC6 and AdC7 were markedly lower. Healthy human adults from the United States had readily detectable circulating T cells recognizing Ad viruses, the levels of which in some individuals were unexpectedly high in response to AdHu26. The magnitude of T-cell responses to AdHu5 correlated with those to AdHu26, suggesting T-cell recognition of conserved epitopes. In mice, all of the different Ad vectors induced CD8(+) T-cell responses that were comparable in their magnitudes and cytokine production profiles. Prime-boost regimens comparing different combinations of Ad vectors failed to indicate that the sequential use of Ad vectors from distinct families resulted in higher immune responses than the use of serologically distinct Ad vectors from the same family. Moreover, the transgene product-specific antibody responses induced by the AdHu26 and AdC vectors were markedly lower than those induced by the AdHu5 vector. AdHu26 vectors and, to a lesser extent, AdC vectors induced more potent Ad-neutralizing antibody responses. These results suggest that the potential of AdHu26 as a vaccine vector may suffer from limitations similar to those found for vectors based on other prevalent human Ads.
Collapse
MESH Headings
- Adenoviridae/classification
- Adenoviridae/genetics
- Adenoviridae/immunology
- Adenoviruses, Human/classification
- Adenoviruses, Human/genetics
- Adenoviruses, Human/immunology
- Adenoviruses, Simian/classification
- Adenoviruses, Simian/genetics
- Adenoviruses, Simian/immunology
- Adult
- Africa South of the Sahara
- Animals
- Antibodies, Neutralizing/blood
- Antibodies, Viral/blood
- CD8-Positive T-Lymphocytes/immunology
- CHO Cells
- Capsid/immunology
- Cell Line
- Cricetinae
- Cricetulus
- Female
- Genetic Vectors
- Green Fluorescent Proteins/genetics
- Green Fluorescent Proteins/metabolism
- Humans
- Mice
- Mice, Inbred BALB C
- Rabies virus/immunology
- Receptors, Virus/metabolism
- Recombinant Proteins/genetics
- Recombinant Proteins/metabolism
- Seroepidemiologic Studies
- Serotyping
- Species Specificity
- Viral Vaccines/genetics
Collapse
Affiliation(s)
- H. Chen
- The Wistar Institute, Philadelphia, Pennsylvania, Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, AIDS Research Unit, National Institute for Communicable Diseases, Johannesburg, South Africa, MRC/UVRI Uganda Research Unit on AIDS, Entebbe, Uganda, Department of Immunology, Tong Ji University, Shanghai, China, Department of Infectious Diseases, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Z. Q. Xiang
- The Wistar Institute, Philadelphia, Pennsylvania, Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, AIDS Research Unit, National Institute for Communicable Diseases, Johannesburg, South Africa, MRC/UVRI Uganda Research Unit on AIDS, Entebbe, Uganda, Department of Immunology, Tong Ji University, Shanghai, China, Department of Infectious Diseases, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Y. Li
- The Wistar Institute, Philadelphia, Pennsylvania, Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, AIDS Research Unit, National Institute for Communicable Diseases, Johannesburg, South Africa, MRC/UVRI Uganda Research Unit on AIDS, Entebbe, Uganda, Department of Immunology, Tong Ji University, Shanghai, China, Department of Infectious Diseases, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - R. K. Kurupati
- The Wistar Institute, Philadelphia, Pennsylvania, Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, AIDS Research Unit, National Institute for Communicable Diseases, Johannesburg, South Africa, MRC/UVRI Uganda Research Unit on AIDS, Entebbe, Uganda, Department of Immunology, Tong Ji University, Shanghai, China, Department of Infectious Diseases, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - B. Jia
- The Wistar Institute, Philadelphia, Pennsylvania, Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, AIDS Research Unit, National Institute for Communicable Diseases, Johannesburg, South Africa, MRC/UVRI Uganda Research Unit on AIDS, Entebbe, Uganda, Department of Immunology, Tong Ji University, Shanghai, China, Department of Infectious Diseases, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - A. Bian
- The Wistar Institute, Philadelphia, Pennsylvania, Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, AIDS Research Unit, National Institute for Communicable Diseases, Johannesburg, South Africa, MRC/UVRI Uganda Research Unit on AIDS, Entebbe, Uganda, Department of Immunology, Tong Ji University, Shanghai, China, Department of Infectious Diseases, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - D. M. Zhou
- The Wistar Institute, Philadelphia, Pennsylvania, Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, AIDS Research Unit, National Institute for Communicable Diseases, Johannesburg, South Africa, MRC/UVRI Uganda Research Unit on AIDS, Entebbe, Uganda, Department of Immunology, Tong Ji University, Shanghai, China, Department of Infectious Diseases, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - N. Hutnick
- The Wistar Institute, Philadelphia, Pennsylvania, Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, AIDS Research Unit, National Institute for Communicable Diseases, Johannesburg, South Africa, MRC/UVRI Uganda Research Unit on AIDS, Entebbe, Uganda, Department of Immunology, Tong Ji University, Shanghai, China, Department of Infectious Diseases, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - S. Yuan
- The Wistar Institute, Philadelphia, Pennsylvania, Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, AIDS Research Unit, National Institute for Communicable Diseases, Johannesburg, South Africa, MRC/UVRI Uganda Research Unit on AIDS, Entebbe, Uganda, Department of Immunology, Tong Ji University, Shanghai, China, Department of Infectious Diseases, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - C. Gray
- The Wistar Institute, Philadelphia, Pennsylvania, Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, AIDS Research Unit, National Institute for Communicable Diseases, Johannesburg, South Africa, MRC/UVRI Uganda Research Unit on AIDS, Entebbe, Uganda, Department of Immunology, Tong Ji University, Shanghai, China, Department of Infectious Diseases, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - J. Serwanga
- The Wistar Institute, Philadelphia, Pennsylvania, Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, AIDS Research Unit, National Institute for Communicable Diseases, Johannesburg, South Africa, MRC/UVRI Uganda Research Unit on AIDS, Entebbe, Uganda, Department of Immunology, Tong Ji University, Shanghai, China, Department of Infectious Diseases, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - B. Auma
- The Wistar Institute, Philadelphia, Pennsylvania, Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, AIDS Research Unit, National Institute for Communicable Diseases, Johannesburg, South Africa, MRC/UVRI Uganda Research Unit on AIDS, Entebbe, Uganda, Department of Immunology, Tong Ji University, Shanghai, China, Department of Infectious Diseases, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - P. Kaleebu
- The Wistar Institute, Philadelphia, Pennsylvania, Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, AIDS Research Unit, National Institute for Communicable Diseases, Johannesburg, South Africa, MRC/UVRI Uganda Research Unit on AIDS, Entebbe, Uganda, Department of Immunology, Tong Ji University, Shanghai, China, Department of Infectious Diseases, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - X. Zhou
- The Wistar Institute, Philadelphia, Pennsylvania, Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, AIDS Research Unit, National Institute for Communicable Diseases, Johannesburg, South Africa, MRC/UVRI Uganda Research Unit on AIDS, Entebbe, Uganda, Department of Immunology, Tong Ji University, Shanghai, China, Department of Infectious Diseases, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - M. R. Betts
- The Wistar Institute, Philadelphia, Pennsylvania, Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, AIDS Research Unit, National Institute for Communicable Diseases, Johannesburg, South Africa, MRC/UVRI Uganda Research Unit on AIDS, Entebbe, Uganda, Department of Immunology, Tong Ji University, Shanghai, China, Department of Infectious Diseases, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - H. C. J. Ertl
- The Wistar Institute, Philadelphia, Pennsylvania, Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, AIDS Research Unit, National Institute for Communicable Diseases, Johannesburg, South Africa, MRC/UVRI Uganda Research Unit on AIDS, Entebbe, Uganda, Department of Immunology, Tong Ji University, Shanghai, China, Department of Infectious Diseases, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
42
|
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
|
43
|
Sharma A, Bangari DS, Tandon M, Hogenesch H, Mittal SK. Evaluation of innate immunity and vector toxicity following inoculation of bovine, porcine or human adenoviral vectors in a mouse model. Virus Res 2010; 153:134-42. [PMID: 20659505 DOI: 10.1016/j.virusres.2010.07.021] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2010] [Revised: 07/15/2010] [Accepted: 07/17/2010] [Indexed: 01/16/2023]
Abstract
Nonhuman adenovirus (Ad) vectors derived from bovine Ad serotype 3 (BAd3) or porcine Ad serotype 3 (PAd3) can circumvent pre-existing immunity against human Ad (HAd). We have previously reported differential transduction of human and nonhuman cells by these Ad vectors, and their distinct receptor usage and biodistribution. To compare the induction of innate immunity, vector toxicity and vector uptake by Kupffer cells (KCs) following intravenous administration of PAd3, BAd3, or HAd5 vectors in mice, we determined mRNA expression levels of proinflammatory chemokines and cytokines, and Toll-like receptors (TLRs) in the liver and spleen. Tissue toxicity of these vectors was assessed by comparing serum levels of liver-specific enzymes, histopathology and Kupffer cell (KC) depletion. Compared to the HAd5 vector, PAd3 and BAd3 vectors were more potent stimulators of innate immune responses as indicated by enhanced mRNA expression of TLRs and proinflammatory chemokines and cytokine genes. Histopathological changes in the liver were most pronounced in HAd5-inoculated mice while BAd3- or PAd3-inoculated mice revealed mild histologic changes that were confined to early time points. Inoculation with HAd5 or PAd3 vectors resulted in a significant (P<0.05) decline of the number of KCs in the liver. Together, these results extend our previous observations regarding distinct in vivo biology of nonhuman and human Ad vectors.
Collapse
Affiliation(s)
- Anurag Sharma
- Department of Comparative Pathobiology, School of Veterinary Medicine, and Bindley Bioscience Center, Purdue University, West Lafayette, IN 47907, USA
| | | | | | | | | |
Collapse
|
44
|
Singh S, Toro H, Tang DC, Briles WE, Yates LM, Kopulos RT, Collisson EW. Non-replicating adenovirus vectors expressing avian influenza virus hemagglutinin and nucleocapsid proteins induce chicken specific effector, memory and effector memory CD8(+) T lymphocytes. Virology 2010; 405:62-9. [PMID: 20557918 DOI: 10.1016/j.virol.2010.05.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2010] [Revised: 04/04/2010] [Accepted: 05/02/2010] [Indexed: 10/19/2022]
Abstract
Avian influenza virus (AIV) specific CD8(+) T lymphocyte responses stimulated by intramuscular administration of an adenovirus (Ad) vector expressing either HA or NP were evaluated in chickens following ex vivo stimulation by non-professional antigen presenting cells. The CD8(+) T lymphocyte responses were AIV specific, MHC-I restricted, and cross-reacted with heterologous H7N2 AIV strain. Specific effector responses, at 10 days post-inoculation (p.i.), were undetectable at 2 weeks p.i., and memory responses were detected from 3 to 8 weeks p.i. Effector memory responses, detected 1 week following a booster inoculation, were significantly greater than the primary responses and, within 7 days, declined to undetectable levels. Inoculation of an Ad-vector expressing human NP resulted in significantly greater MHC restricted, activation of CD8(+) T cell responses specific for AIV. Decreases in all responses with time were most dramatic with maximum activation of T cells as observed following effector and effector memory responses.
Collapse
Affiliation(s)
- Shailbala Singh
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Texas A&M University, College Station, TX 77843, USA
| | | | | | | | | | | | | |
Collapse
|
45
|
Steitz J, Barlow PG, Hossain J, Kim E, Okada K, Kenniston T, Rea S, Donis RO, Gambotto A. A candidate H1N1 pandemic influenza vaccine elicits protective immunity in mice. PLoS One 2010; 5:e10492. [PMID: 20463955 PMCID: PMC2864737 DOI: 10.1371/journal.pone.0010492] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2010] [Accepted: 04/08/2010] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND In 2009 a new pandemic disease appeared and spread globally. The recent emergence of the pandemic influenza virus H1N1 first isolated in Mexico and USA raised concerns about vaccine availability. We here report our development of an adenovirus-based influenza H1N1 vaccine tested for immunogenicity and efficacy to confer protection in animal model. METHODS We generated two adenovirus(Ad5)-based influenza vaccine candidates encoding the wildtype or a codon-optimized hemagglutinin antigen (HA) from the recently emerged swine influenza isolate A/California/04/2009 (H1N1)pdm. After verification of antigen expression, immunogenicity of the vaccine candidates were tested in a mouse model using dose escalations for subcutaneous immunization. Sera of immunized animals were tested in microneutalization and hemagglutination inhibition assays for the presence of HA-specific antibodies. HA-specific T-cells were measured in IFNgamma Elispot assays. The efficiency of the influenza vaccine candidates were evaluated in a challenge model by measuring viral titer in lung and nasal turbinate 3 days after inoculation of a homologous H1N1 virus. CONCLUSIONS/SIGNIFICANCE A single immunization resulted in robust cellular and humoral immune response. Remarkably, the intensity of the immune response was substantially enhanced with codon-optimized antigen, indicating the benefit of manipulating the genetic code of HA antigens in the context of recombinant influenza vaccine design. These results highlight the value of advanced technologies in vaccine development and deployment in response to infections with pandemic potential. Our study emphasizes the potential of an adenoviral-based influenza vaccine platform with the benefits of speed of manufacture and efficacy of a single dose immunization.
Collapse
Affiliation(s)
- Julia Steitz
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Peter G. Barlow
- Molecular Virology and Vaccines Branch, Influenza Division, National Center for Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Jaber Hossain
- Molecular Virology and Vaccines Branch, Influenza Division, National Center for Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Eun Kim
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Kaori Okada
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Tom Kenniston
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Sheri Rea
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Ruben O. Donis
- Molecular Virology and Vaccines Branch, Influenza Division, National Center for Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Andrea Gambotto
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- Division of Infectious Diseases, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| |
Collapse
|
46
|
Avian influenza pandemic preparedness: developing prepandemic and pandemic vaccines against a moving target. Expert Rev Mol Med 2010; 12:e14. [PMID: 20426889 DOI: 10.1017/s1462399410001432] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The unprecedented global spread of highly pathogenic avian H5N1 influenza viruses within the past ten years and their extreme lethality to poultry and humans has underscored their potential to cause an influenza pandemic. Combating the threat of an impending H5N1 influenza pandemic will require a combination of pharmaceutical and nonpharmaceutical intervention strategies. The emergence of the H1N1 pandemic in 2009 emphasised the unpredictable nature of a pandemic influenza. Undoubtedly, vaccines offer the most viable means to combat a pandemic threat. Current egg-based influenza vaccine manufacturing strategies are unlikely to be able to cater to the huge, rapid global demand because of the anticipated scarcity of embryonated eggs in an avian influenza pandemic and other factors associated with the vaccine production process. Therefore, alternative, egg-independent vaccine manufacturing strategies should be evaluated to supplement the traditional egg-derived influenza vaccine manufacturing. Furthermore, evaluation of dose-sparing strategies that offer protection with a reduced antigen dose will be critical for pandemic influenza preparedness. Development of new antiviral therapeutics and other, nonpharmaceutical intervention strategies will further supplement pandemic preparedness. This review highlights the current status of egg-dependent and egg-independent strategies against an avian influenza pandemic.
Collapse
|
47
|
Evaluation of cross-reactive cell-mediated immune responses among human, bovine and porcine adenoviruses. Gene Ther 2010; 17:634-42. [PMID: 20164856 PMCID: PMC2869393 DOI: 10.1038/gt.2010.1] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The absence of preexisting immunity against porcine adenovirus (Ad) serotype 3 (PAd3) and bovine Ad serotype 3 (BAd3) in humans makes them attractive alternatives to human Ad serotype 5 (HAd5) vectors. To determine whether there is significant cross-reactivity among HAd5, BAd3, and PAd3 at the level of cell-mediated immune responses, BALB/c mice were inoculated intraperitoneally with wild type (WT) or replication-defective (RD) HAd5, BAd3, or PAd3. Thirty-five days after the first inoculation, cross-reactive CD8+ cytotoxic T cells, as well as CD4+ Th1- and Th2-helper T cells, in the spleen were analyzed by ELISPOT, flow cytometry and cytotoxic T lymphocyte (CTL) assays. Virus neutralization assays were used to evaluate humoral cross-reactivity. CD8+ or CD4+ T cells primed with WT or RD HAd5, PAd3, or BAd3 demonstrated significant (P <0.005) reactivity with homologous Ad antigens, whereas, only minimal cross-reactivity was observed upon stimulation with heterologous Ad antigens. Ad-neutralizing antibodies were found to be homologous Ad-specific. Overall, these results suggest that there is no significant immunological cross-reactivity among HAd5, BAd3, and PAd3, thereby supporting the rationale for the use of BAd3 and PAd3 as alternative HAd vectors to circumvent anti-HAd immunity in humans.
Collapse
|
48
|
Pandey A, Singh N, Sambhara S, Mittal SK. Egg-independent vaccine strategies for highly pathogenic H5N1 influenza viruses. HUMAN VACCINES 2010; 6:178-88. [PMID: 19875936 PMCID: PMC2888842 DOI: 10.4161/hv.6.2.9899] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The emergence of a highly pathogenic H5N1 influenza virus in Hong Kong in 1997 and the subsequent appearance of other H5N1 strains and their spread to several countries in southeast Asia, Africa, the Middle East and Europe has evoked fear of a global influenza pandemic. Vaccines offer the best hope to combat the threat of an influenza pandemic. However, the global demand for a pandemic vaccine cannot be fulfilled by the current egg-based vaccine manufacturing strategies, thus creating a need to explore alternative technologies for vaccine production and delivery. Several egg-independent vaccine approaches such as cell culture-derived whole virus or subvirion vaccines, recombinant protein-based vaccines, virus-like particle (VLP) vaccines, DNA vaccines and viral vector-based vaccines are currently being investigated and appear promising both in preclinical and clinical studies. The present review will highlight the various egg-independent alternative vaccine approaches for pandemic influenza.
Collapse
Affiliation(s)
| | | | | | - Suresh K. Mittal
- Correspondence: Suresh K. Mittal, Department of Comparative Pathobiology, School of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA, Tel: 765-496-2894, Fax: 765-494-9830, , Suryaprakash Sambhara, Influenza Division, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA, Tel: 404-639-3800, Fax: 404-639-5180,
| |
Collapse
|
49
|
A complex adenovirus-vectored vaccine against Rift Valley fever virus protects mice against lethal infection in the presence of preexisting vector immunity. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2009; 16:1624-32. [PMID: 19776190 DOI: 10.1128/cvi.00182-09] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Rift Valley fever virus (RVFV) has been cited as a potential biological-weapon threat due to the serious and fatal disease it causes in humans and animals and the fact that this mosquito-borne virus can be lethal in an aerosolized form. Current human and veterinary vaccines against RVFV, however, are outdated, inefficient, and unsafe. We have incorporated the RVFV glycoprotein genes into a nonreplicating complex adenovirus (CAdVax) vector platform to develop a novel RVFV vaccine. Mice vaccinated with the CAdVax-based vaccine produced potent humoral immune responses and were protected against lethal RVFV infection. Additionally, protection was elicited in mice despite preexisting immunity to the adenovirus vector.
Collapse
|
50
|
Nayak B, Rout SN, Kumar S, Khalil MS, Fouda MM, Ahmed LE, Earhart KC, Perez DR, Collins PL, Samal SK. Immunization of chickens with Newcastle disease virus expressing H5 hemagglutinin protects against highly pathogenic H5N1 avian influenza viruses. PLoS One 2009; 4:e6509. [PMID: 19654873 PMCID: PMC2716524 DOI: 10.1371/journal.pone.0006509] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2009] [Accepted: 07/02/2009] [Indexed: 11/18/2022] Open
Abstract
Background Highly-pathogenic avian influenza virus (HPAIV) and Newcastle disease virus (NDV) are the two most important poultry viruses in the world. Natural low-virulence NDV strains have been used as vaccines over the past 70 years with proven track records. We have previously developed a reverse genetics system to produce low-virulent NDV vaccine strain LaSota from cloned cDNA. This system allows us to use NDV as a vaccine vector for other avian pathogens. Methodology/Principal Finding Here, we constructed two recombinant NDVs (rNDVs) each of which expresses the hemagglutinin (HA) gene of HPAIV H5N1strain A/Vietnam/1203/2004 from an added gene. In one, rNDV (rNDV-HA), the open reading frame (ORF) of HA gene was expressed without modification. In the second, rNDV (rNDV-HAF), the ORF was modified so that the transmembrane and cytoplasmic domains of the encoded HA gene were replaced with those of the NDV F protein. The insertion of either version of the HA ORF did not increase the virulence of the rNDV vector. The HA protein was found to be incorporated into the envelopes of both rNDV-HA and rNDV-HAF. However, there was an enhanced incorporation of the HA protein in rNDV-HAF. Chickens immunized with a single dose of either rNDV-HA or rNDV-HAF induced a high titer of HPAIV H5-specific antibodies and were completely protected against challenge with NDV as well as lethal challenges of both homologous and heterologous HPAIV H5N1. Conclusion and Significance Our results suggest that these chimeric viruses have potential as safe and effective bivalent vaccines against NDV and. HPAIV. These vaccines will be convenient and affordable, which will be highly beneficial to the poultry industry. Furthermore, immunization with these vaccines will permit serological differentiation of vaccinated and avian influenza field virus infected animals.
Collapse
Affiliation(s)
- Baibaswata Nayak
- Virginia-Maryland Regional College of Veterinary Medicine, University of Maryland, College Park, Maryland, United States of America
| | - Subrat N. Rout
- Virginia-Maryland Regional College of Veterinary Medicine, University of Maryland, College Park, Maryland, United States of America
| | - Sachin Kumar
- Virginia-Maryland Regional College of Veterinary Medicine, University of Maryland, College Park, Maryland, United States of America
| | | | | | | | | | - Daniel R. Perez
- Virginia-Maryland Regional College of Veterinary Medicine, University of Maryland, College Park, Maryland, United States of America
| | - Peter L. Collins
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, United States of America
| | - Siba K. Samal
- Virginia-Maryland Regional College of Veterinary Medicine, University of Maryland, College Park, Maryland, United States of America
- * E-mail:
| |
Collapse
|