1
|
Murala MST, Gairola V, Sayedahmed EE, Mittal SK. Next-Generation Adenoviral Vector-Based Vaccines for Severe Acute Respiratory Syndrome Coronavirus-2. Vaccines (Basel) 2025; 13:406. [PMID: 40333307 PMCID: PMC12031563 DOI: 10.3390/vaccines13040406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Revised: 04/08/2025] [Accepted: 04/11/2025] [Indexed: 05/09/2025] Open
Abstract
This review systematically revises adenovirus (Ad) biology, vector structure, immune responses, and currently available Ad vector COVID-19 vaccines. It analyzes the challenges associated with the Ad vector-based vaccines, including preexisting vector immunity and other side effects. Moreover, this review explores novel and innovative strategies to overcome these constraints for developing next-generation vaccines for broad protection to cover emerging SARS-CoV-2 variants. The future refinement of Ad vaccine platforms will be pivotal in achieving durable immunity against emerging variants for global preparedness.
Collapse
Affiliation(s)
| | | | | | - Suresh K. Mittal
- Department of Comparative Pathobiology, Purdue Institute of Inflammation, Immunology and Infectious Disease, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907-2027, USA; (M.S.T.M.); (V.G.); (E.E.S.)
| |
Collapse
|
2
|
Wang WC, Sayedahmed EE, Alhashimi M, Elkashif A, Gairola V, Murala MST, Sambhara S, Mittal SK. Adenoviral Vector-Based Vaccine Expressing Hemagglutinin Stem Region with Autophagy-Inducing Peptide Confers Cross-Protection Against Group 1 and 2 Influenza A Viruses. Vaccines (Basel) 2025; 13:95. [PMID: 39852874 PMCID: PMC11769558 DOI: 10.3390/vaccines13010095] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 01/07/2025] [Accepted: 01/14/2025] [Indexed: 01/26/2025] Open
Abstract
Background/Objectives: An effective universal influenza vaccine is urgently needed to overcome the limitations of current seasonal influenza vaccines, which are ineffective against mismatched strains and unable to protect against pandemic influenza. Methods: In this study, bovine and human adenoviral vector-based vaccine platforms were utilized to express various combinations of antigens. These included the H5N1 hemagglutinin (HA) stem region or HA2, the extracellular domain of matrix protein 2 of influenza A virus, HA signal peptide (SP), trimerization domain, excretory peptide, and the autophagy-inducing peptide C5 (AIP-C5). The goal was to identify the optimal combination for enhanced immune responses and cross-protection. Mice were immunized using a prime-boost strategy with heterologous adenoviral (Ad) vectors. Results: The heterologous Ad vectors induced robust HA stem-specific humoral and cellular immune responses in the immunized mice. Among the tested combinations, Ad vectors expressing SP + HA stem + AIP-C5 conferred significant protection against group 1 (H1N1 and H5N1) and group 2 (H3N2) influenza A viruses. This protection was demonstrated by lower lung viral titers and reduced morbidity and mortality. Conclusions: The findings support further investigation of heterologous Ad vaccine platforms expressing SP + HA stem + AIP-C5. This combination shows promise as a potential universal influenza vaccine, providing broader protection against influenza A viruses.
Collapse
Affiliation(s)
- Wen-Chien Wang
- Department of Comparative Pathobiology, Purdue Institute of Inflammation, Immunology and Infectious Disease, College of Veterinary Medicine, Purdue University, 625 Harrison St., West Lafayette, IN 47907, USA; (W.-C.W.); (E.E.S.); (M.A.); (A.E.); (V.G.); (M.S.T.M.)
| | - Ekramy E. Sayedahmed
- Department of Comparative Pathobiology, Purdue Institute of Inflammation, Immunology and Infectious Disease, College of Veterinary Medicine, Purdue University, 625 Harrison St., West Lafayette, IN 47907, USA; (W.-C.W.); (E.E.S.); (M.A.); (A.E.); (V.G.); (M.S.T.M.)
| | - Marwa Alhashimi
- Department of Comparative Pathobiology, Purdue Institute of Inflammation, Immunology and Infectious Disease, College of Veterinary Medicine, Purdue University, 625 Harrison St., West Lafayette, IN 47907, USA; (W.-C.W.); (E.E.S.); (M.A.); (A.E.); (V.G.); (M.S.T.M.)
| | - Ahmed Elkashif
- Department of Comparative Pathobiology, Purdue Institute of Inflammation, Immunology and Infectious Disease, College of Veterinary Medicine, Purdue University, 625 Harrison St., West Lafayette, IN 47907, USA; (W.-C.W.); (E.E.S.); (M.A.); (A.E.); (V.G.); (M.S.T.M.)
| | - Vivek Gairola
- Department of Comparative Pathobiology, Purdue Institute of Inflammation, Immunology and Infectious Disease, College of Veterinary Medicine, Purdue University, 625 Harrison St., West Lafayette, IN 47907, USA; (W.-C.W.); (E.E.S.); (M.A.); (A.E.); (V.G.); (M.S.T.M.)
| | - Muralimanohara S. T. Murala
- Department of Comparative Pathobiology, Purdue Institute of Inflammation, Immunology and Infectious Disease, College of Veterinary Medicine, Purdue University, 625 Harrison St., West Lafayette, IN 47907, USA; (W.-C.W.); (E.E.S.); (M.A.); (A.E.); (V.G.); (M.S.T.M.)
| | - Suryaprakash Sambhara
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| | - Suresh K. Mittal
- Department of Comparative Pathobiology, Purdue Institute of Inflammation, Immunology and Infectious Disease, College of Veterinary Medicine, Purdue University, 625 Harrison St., West Lafayette, IN 47907, USA; (W.-C.W.); (E.E.S.); (M.A.); (A.E.); (V.G.); (M.S.T.M.)
| |
Collapse
|
3
|
Xie CZ, Zhang P, Wang Z, Tao YM, Cui ZD, Nan FL, Zhang FC, Ren YX, Zhang H, Lu HJ. Construction and immunological evaluation of recombinant adenovirus vaccines of new novel NADC34-PRRSV strains in pigs. Front Vet Sci 2024; 11:1503526. [PMID: 39742318 PMCID: PMC11685224 DOI: 10.3389/fvets.2024.1503526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 11/25/2024] [Indexed: 01/03/2025] Open
Abstract
Introduction Porcine reproductive and respiratory syndrome virus (PRRSV) causes reproductive and respiratory diseases in sow herds and piglets. The emergence of ORF5 RFLP 1-7-4-like (NADC34-like) PRRSV strain in China has brought a new round of challenges to PRRSV prevention. Methods In addition, recombinant adenovirus vaccine candidates against the newly emerged NADC34-like strain were constructed in the study; the immunogenicity of the vaccine was investigated in piglets. After inoculation with PRRSV recombinant adenovirus, specific antibodies, neutralizing antibodies, and levels of IFN-γ and IL-4 cytokines were detected in serum. Results Thirty-five days after immunization, the levels of IFN-γ and IL-4 cytokines in the pac-Ad5-34-GP3, pac-Ad5-34-GP5, and pac-Ad5-34-GP35 experimental groups were significantly higher (p < 0.05) than those of the PBS and the adenovirus group. All vaccines can cause corresponding Th1 and Th2 immune responses based on animal experimental results. After the challenge, no obvious clinical symptoms were observed in the immune groups compared with the control group, vaccinated animals could reduce the occurrence of viremia, and the occurrence of viremia was alleviated, with no obvious pathological changes in the lungs, indicating that recombinant adenovirus vaccine could provide a good protective immunity and produce a good humoral and cellular immune response at the same time. Discussion It shows that the recombinant adenovirus vaccine group has better protection against the virus. Provide vaccine reserve and theoretical support for the emergence of new PRRSV subtypes in China.
Collapse
Affiliation(s)
- Chang-zhan Xie
- Institute of Urban Agriculture, Chengdu National Agricultural Science & Technology Center, Chinese Academy of Agricultural Sciences, Chengdu, China
- Key Laboratory of Special Animal Epidemic Disease, Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun, China
- Changchun Veterinary Research Institute, State Key Laboratory of Pathogen and Biosecurity, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Ping Zhang
- Institute of Urban Agriculture, Chengdu National Agricultural Science & Technology Center, Chinese Academy of Agricultural Sciences, Chengdu, China
- Key Laboratory of Special Animal Epidemic Disease, Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun, China
- Changchun Veterinary Research Institute, State Key Laboratory of Pathogen and Biosecurity, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Zheng Wang
- Changchun Veterinary Research Institute, State Key Laboratory of Pathogen and Biosecurity, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Yi-mo Tao
- Changchun Veterinary Research Institute, State Key Laboratory of Pathogen and Biosecurity, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Zhuo-dong Cui
- Changchun Veterinary Research Institute, State Key Laboratory of Pathogen and Biosecurity, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Fu-long Nan
- Changchun Veterinary Research Institute, State Key Laboratory of Pathogen and Biosecurity, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Fu-chao Zhang
- College of Animal Science and Veterinary Medicine, Southwest Minzu University, Chengdu, China
| | - Yun-xin Ren
- College of Animal Science and Veterinary Medicine, Southwest Minzu University, Chengdu, China
| | - He Zhang
- Changchun Veterinary Research Institute, State Key Laboratory of Pathogen and Biosecurity, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Hui-jun Lu
- Changchun Veterinary Research Institute, State Key Laboratory of Pathogen and Biosecurity, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Chinese Academy of Agricultural Sciences, Changchun, China
| |
Collapse
|
4
|
Zhang P, Luo S, Zou P, Deng Q, Wang C, Li J, Cai P, Zhang L, Li C, Li T. A novel simian adenovirus-vectored COVID-19 vaccine elicits effective mucosal and systemic immunity in mice by intranasal and intramuscular vaccination regimens. Microbiol Spectr 2023; 11:e0179423. [PMID: 37877750 PMCID: PMC10715068 DOI: 10.1128/spectrum.01794-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 09/19/2023] [Indexed: 10/26/2023] Open
Abstract
IMPORTANCE The essential goal of vaccination is to generate potent and long-term protection against diseases. Several factors including vaccine vector, delivery route, and boosting regimen influence the outcome of prime-boost immunization approaches. The immunization regimens by constructing a novel simian adenovirus-vectored COVID-19 vaccine and employing combination of intranasal and intramuscular inoculations could elicit mucosal neutralizing antibodies against five mutant strains in the respiratory tract and strong systemic immunity. Immune protection could last for more than 32 weeks. Vectored vaccine construction and immunization regimens have positively impacted respiratory disease prevention.
Collapse
Affiliation(s)
- Panli Zhang
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Guangzhou Bai Rui Kang (BRK) Biological Science and Technology Limited Company, Guangzhou , China
| | - Shengxue Luo
- Department of Pediatrics, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Peng Zou
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Guangzhou Bai Rui Kang (BRK) Biological Science and Technology Limited Company, Guangzhou , China
| | - Qitao Deng
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Guangzhou Bai Rui Kang (BRK) Biological Science and Technology Limited Company, Guangzhou , China
| | - Cong Wang
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Guangzhou Bai Rui Kang (BRK) Biological Science and Technology Limited Company, Guangzhou , China
| | - Jinfeng Li
- Shenzhen Bao'an District Central Blood Station, Shenzhen, China
| | - Peiqiao Cai
- Department of Bioengineering, School of Medicine and College of Engineering, University of Washington, Seattle, Washington, USA
| | - Ling Zhang
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Chengyao Li
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Tingting Li
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| |
Collapse
|
5
|
Wang WC, Sayedahmed EE, Mittal SK. Significance of Preexisting Vector Immunity and Activation of Innate Responses for Adenoviral Vector-Based Therapy. Viruses 2022; 14:v14122727. [PMID: 36560730 PMCID: PMC9787786 DOI: 10.3390/v14122727] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/01/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022] Open
Abstract
An adenoviral (AdV)-based vector system is a promising platform for vaccine development and gene therapy applications. Administration of an AdV vector elicits robust innate immunity, leading to the development of humoral and cellular immune responses against the vector and the transgene antigen, if applicable. The use of high doses (1011-1013 virus particles) of an AdV vector, especially for gene therapy applications, could lead to vector toxicity due to excessive levels of innate immune responses, vector interactions with blood factors, or high levels of vector transduction in the liver and spleen. Additionally, the high prevalence of AdV infections in humans or the first inoculation with the AdV vector result in the development of vector-specific immune responses, popularly known as preexisting vector immunity. It significantly reduces the vector efficiency following the use of an AdV vector that is prone to preexisting vector immunity. Several approaches have been developed to overcome this problem. The utilization of rare human AdV types or nonhuman AdVs is the primary strategy to evade preexisting vector immunity. The use of heterologous viral vectors, capsid modification, and vector encapsulation are alternative methods to evade vector immunity. The vectors can be optimized for clinical applications with comprehensive knowledge of AdV vector immunity, toxicity, and circumvention strategies.
Collapse
|
6
|
Huang S, Zhu Y, Zhang L, Zhang Z. Recent Advances in Delivery Systems for Genetic and Other Novel Vaccines. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2107946. [PMID: 34914144 DOI: 10.1002/adma.202107946] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 12/11/2021] [Indexed: 06/14/2023]
Abstract
Vaccination is one of the most successful and cost-effective prophylactic measures against diseases, especially infectious diseases including smallpox and polio. However, the development of effective prophylactic or therapeutic vaccines for other diseases such as cancer remains challenging. This is often due to the imprecise control of vaccine activity in vivo which leads to insufficient/inappropriate immune responses or short immune memory. The development of new vaccine types in recent decades has created the potential for improving the protective potency against these diseases. Genetic and subunit vaccines are two major categories of these emerging vaccines. Owing to their nature, they rely heavily on delivery systems with various functions, such as effective cargo protection, immunogenicity enhancement, targeted delivery, sustained release of antigens, selective activation of humoral and/or cellular immune responses against specific antigens, and reduced adverse effects. Therefore, vaccine delivery systems may significantly affect the final outcome of genetic and other novel vaccines and are vital for their development. This review introduces these studies based on their research emphasis on functional design or administration route optimization, presents recent progress, and discusses features of new vaccine delivery systems, providing an overview of this field.
Collapse
Affiliation(s)
- Shiqi Huang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, College of Polymer Science and Engineering, Sichuan University, Chengdu, 610041, P. R. China
| | - Yining Zhu
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, College of Polymer Science and Engineering, Sichuan University, Chengdu, 610041, P. R. China
| | - Ling Zhang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, College of Polymer Science and Engineering, Sichuan University, Chengdu, 610041, P. R. China
| | - Zhirong Zhang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, College of Polymer Science and Engineering, Sichuan University, Chengdu, 610041, P. R. China
| |
Collapse
|
7
|
Wang R, Pan W, Jin L, Huang W, Li Y, Wu D, Gao C, Ma D, Liao S. Human papillomavirus vaccine against cervical cancer: Opportunity and challenge. Cancer Lett 2020; 471:88-102. [DOI: 10.1016/j.canlet.2019.11.039] [Citation(s) in RCA: 258] [Impact Index Per Article: 51.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 11/28/2019] [Accepted: 11/30/2019] [Indexed: 12/20/2022]
|
8
|
Stylianou E, Paul MJ, Reljic R, McShane H. Mucosal delivery of tuberculosis vaccines: a review of current approaches and challenges. Expert Rev Vaccines 2019; 18:1271-1284. [PMID: 31876199 PMCID: PMC6961305 DOI: 10.1080/14760584.2019.1692657] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Introduction: Tuberculosis (TB) remains a major health threat and it is now clear that the current vaccine, BCG, is unable to arrest the global TB epidemic. A new vaccine is needed to either replace or boost BCG so that a better level of protection could be achieved. The route of entry of Mycobacterium tuberculosis, the causative organism, is via inhalation making TB primarily a respiratory disease. There is therefore good reason to hypothesize that a mucosally delivered vaccine against TB could be more effective than one delivered via the systemic route. Areas covered: This review summarizes the progress that has been made in the area of TB mucosal vaccines in the last few years. It highlights some of the strengths and shortcomings of the published evidence and aims to discuss immunological and practical considerations in the development of mucosal vaccines. Expert opinion: There is a growing body of evidence that the mucosal approach to vaccination against TB is feasible and should be pursued. However, further key studies are necessary to both improve our understanding of the protective immune mechanisms operating in the mucosa and the technical aspects of aerosolized delivery, before such a vaccine could become a feasible, deployable strategy.
Collapse
Affiliation(s)
- Elena Stylianou
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Matthew J Paul
- Institute for Infection and Immunity, St George's University of London, Tooting, London, UK
| | - Rajko Reljic
- Institute for Infection and Immunity, St George's University of London, Tooting, London, UK
| | - Helen McShane
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
9
|
Sharma PK, Dmitriev IP, Kashentseva EA, Raes G, Li L, Kim SW, Lu ZH, Arbeit JM, Fleming TP, Kaliberov SA, Goedegebuure SP, Curiel DT, Gillanders WE. Development of an adenovirus vector vaccine platform for targeting dendritic cells. Cancer Gene Ther 2018; 25:27-38. [PMID: 29242639 PMCID: PMC5972836 DOI: 10.1038/s41417-017-0002-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 08/04/2017] [Accepted: 08/07/2017] [Indexed: 12/16/2022]
Abstract
Adenoviral (Ad) vector vaccines represent one of the most promising modern vaccine platforms, and Ad vector vaccines are currently being investigated in human clinical trials for infectious disease and cancer. Our studies have shown that specific targeting of adenovirus to dendritic cells dramatically enhanced vaccine efficacy. However, this was achieved using a molecular adapter, thereby necessitating a two component vector approach. To address the mandates of clinical translation of our strategy, we here sought to accomplish the goal of DC targeting with a single-component adenovirus vector approach. To redirect the specificity of Ad vector vaccines, we replaced the Ad fiber knob with fiber-fibritin chimeras fused to DC1.8, a single-domain antibody (sdAb) specific for murine immature DC. We engineered a fiber-fibritin-sdAb chimeric molecule using the coding sequence for DC1.8, and then replaced the native Ad5 fiber knob sequence by homologous recombination. The resulting Ad5 virus, Ad5FF1.8, expresses the chimeric fiber-fibritin sdAb chimera. Infection with Ad5FF1.8 dramatically enhances transgene expression in DC2.4 dendritic cells compared with infection with native Ad5. Ad5FF1.8 infection of bone marrow-derived DC demonstrates that Ad5FF1.8 selectively infects immature DC consistent with the known specificity of DC1.8. Thus, sdAb can be used to selectively redirect the tropism of Ad5 vector vaccines, providing the opportunity to engineer Ad vector vaccines that are specifically targeted to DC, or specific DC subsets.
Collapse
Affiliation(s)
- Piyush K Sharma
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Igor P Dmitriev
- Cancer Biology Division, Biologic Therapeutics Center, Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Elena A Kashentseva
- Cancer Biology Division, Biologic Therapeutics Center, Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Geert Raes
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
- VIB Center for Inflammation Research, Myeloid Cell Immunology Laboratory, Brussels, Belgium
| | - Lijin Li
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Samuel W Kim
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Zhi-Hong Lu
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Jeffrey M Arbeit
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
- The Alvin J. Siteman Cancer Center, Barnes-Jewish Hospital and Washington University School of Medicine, St. Louis, MO, USA
| | - Timothy P Fleming
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
- The Alvin J. Siteman Cancer Center, Barnes-Jewish Hospital and Washington University School of Medicine, St. Louis, MO, USA
| | - Sergey A Kaliberov
- Cancer Biology Division, Biologic Therapeutics Center, Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - S Peter Goedegebuure
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
- The Alvin J. Siteman Cancer Center, Barnes-Jewish Hospital and Washington University School of Medicine, St. Louis, MO, USA
| | - David T Curiel
- Cancer Biology Division, Biologic Therapeutics Center, Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, USA.
| | - William E Gillanders
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
- The Alvin J. Siteman Cancer Center, Barnes-Jewish Hospital and Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
10
|
Fonseca JA, McCaffery JN, Kashentseva E, Singh B, Dmitriev IP, Curiel DT, Moreno A. A prime-boost immunization regimen based on a simian adenovirus 36 vectored multi-stage malaria vaccine induces protective immunity in mice. Vaccine 2017; 35:3239-3248. [PMID: 28483199 PMCID: PMC5522619 DOI: 10.1016/j.vaccine.2017.04.062] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Revised: 04/20/2017] [Accepted: 04/21/2017] [Indexed: 12/22/2022]
Abstract
Malaria remains a considerable burden on public health. In 2015, the WHO estimates there were 212 million malaria cases causing nearly 429,000 deaths globally. A highly effective malaria vaccine is needed to reduce the burden of this disease. We have developed an experimental vaccine candidate (PyCMP) based on pre-erythrocytic (CSP) and erythrocytic (MSP1) stage antigens derived from the rodent malaria parasite P. yoelii. Our protein-based vaccine construct induces protective antibodies and CD4+ T cell responses. Based on evidence that viral vectors increase CD8+ T cell-mediated immunity, we also have tested heterologous prime-boost immunization regimens that included human adenovirus serotype 5 vector (Ad5), obtaining protective CD8+ T cell responses. While Ad5 is commonly used for vaccine studies, the high prevalence of pre-existing immunity to Ad5 severely compromises its utility. Here, we report the use of the novel simian adenovirus 36 (SAd36) as a candidate for a vectored malaria vaccine since this virus is not known to infect humans, and it is not neutralized by anti-Ad5 antibodies. Our study shows that the recombinant SAd36PyCMP can enhance specific CD8+ T cell response and elicit similar antibody titers when compared to an immunization regimen including the recombinant Ad5PyCMP. The robust immune responses induced by SAd36PyCMP are translated into a lower parasite load following P. yoelii infectious challenge when compared to mice immunized with Ad5PyCMP.
Collapse
Affiliation(s)
- Jairo A Fonseca
- Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, 954 Gatewood Road, Atlanta, GA 30329, United States; Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30307, United States
| | - Jessica N McCaffery
- Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, 954 Gatewood Road, Atlanta, GA 30329, United States
| | - Elena Kashentseva
- Cancer Biology Division, Department of Radiation Oncology, Washington University School of Medicine, 660 S. Euclid Ave., 4511 Forest Park Blvd, St. Louis, MO 63108, United States
| | - Balwan Singh
- Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, 954 Gatewood Road, Atlanta, GA 30329, United States
| | - Igor P Dmitriev
- Cancer Biology Division, Department of Radiation Oncology, Washington University School of Medicine, 660 S. Euclid Ave., 4511 Forest Park Blvd, St. Louis, MO 63108, United States
| | - David T Curiel
- Cancer Biology Division, Department of Radiation Oncology, Washington University School of Medicine, 660 S. Euclid Ave., 4511 Forest Park Blvd, St. Louis, MO 63108, United States
| | - Alberto Moreno
- Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, 954 Gatewood Road, Atlanta, GA 30329, United States; Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30307, United States.
| |
Collapse
|
11
|
Engineering intravaginal vaccines to overcome mucosal and epithelial barriers. Biomaterials 2017; 128:8-18. [PMID: 28285195 DOI: 10.1016/j.biomaterials.2017.03.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 03/03/2017] [Accepted: 03/06/2017] [Indexed: 11/22/2022]
Abstract
The mucosal surface of the vagina is a primary human immunodeficiency virus (HIV) entry portal, making it an attractive site for HIV vaccination. However, HIV vaccines based on recombinant adenovirus (rAd) do not efficiently cross the mucus layers or underlying epithelium of the vagina. Here we designed nanocomplexes of rAd particles coated with (1) the polyethylene glycol derivative APS to provide a hydrophilic surface that would prevent entrapment in the hydrophobic mucus, and (2) the cell-penetrating peptide TAT to improve transduction efficiency. The optimized rAd-TAT-APS nanocomplexes could achieve the balance of effective mucus-penetrating and cellular transduction. Intravaginal delivery of rAd-TAT-APS encoding HIVgag p24 into mice strongly enhanced HIVgag-specific systemic and mucosal immune responses. This rAd-TAT-APS system may allow effective vaginal delivery of vaccines against HIV and other infectious agents.
Collapse
|
12
|
Crosby CM, Matchett WE, Anguiano-Zarate SS, Parks CA, Weaver EA, Pease LR, Webby RJ, Barry MA. Replicating Single-Cycle Adenovirus Vectors Generate Amplified Influenza Vaccine Responses. J Virol 2017; 91:e00720-16. [PMID: 27807231 PMCID: PMC5215357 DOI: 10.1128/jvi.00720-16] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2016] [Accepted: 10/22/2016] [Indexed: 12/14/2022] Open
Abstract
Head-to-head comparisons of conventional influenza vaccines with adenovirus (Ad) gene-based vaccines demonstrated that these viral vectors can mediate more potent protection against influenza virus infection in animal models. In most cases, Ad vaccines are engineered to be replication-defective (RD-Ad) vectors. In contrast, replication-competent Ad (RC-Ad) vaccines are markedly more potent but risk causing adenovirus diseases in vaccine recipients and health care workers. To harness antigen gene replication but avoid production of infectious virions, we developed "single-cycle" adenovirus (SC-Ad) vectors. Previous work demonstrated that SC-Ads amplify transgene expression 100-fold and produce markedly stronger and more persistent immune responses than RD-Ad vectors in Syrian hamsters and rhesus macaques. To test them as potential vaccines, we engineered RD and SC versions of adenovirus serotype 6 (Ad6) to express the hemagglutinin (HA) gene from influenza A/PR/8/34 virus. We show here that it takes approximately 33 times less SC-Ad6 than RD-Ad6 to produce equal amounts of HA antigen in vitro SC-Ad produced markedly higher HA binding and hemagglutination inhibition (HAI) titers than RD-Ad in Syrian hamsters. SC-Ad-vaccinated cotton rats had markedly lower influenza titers than RD-Ad-vaccinated animals after challenge with influenza A/PR/8/34 virus. These data suggest that SC-Ads may be more potent vaccine platforms than conventional RD-Ad vectors and may have utility as "needle-free" mucosal vaccines. IMPORTANCE Most adenovirus vaccines that are being tested are replication-defective adenoviruses (RD-Ads). This work describes testing newer single-cycle adenovirus (SC-Ad) vectors that replicate transgenes to amplify protein production and immune responses. We show that SC-Ads generate markedly more influenza virus hemagglutinin protein and require substantially less vector to generate the same immune responses as RD-Ad vectors. SC-Ads therefore hold promise to be more potent vectors and vaccines than current RD-Ad vectors.
Collapse
Affiliation(s)
- Catherine M Crosby
- Department of Medicine, Division of Infectious Diseases, Mayo Clinic, Rochester, Minnesota, USA
- Virology and Gene Therapy Graduate Program, Mayo Clinic, Rochester, Minnesota, USA
| | - William E Matchett
- Virology and Gene Therapy Graduate Program, Mayo Clinic, Rochester, Minnesota, USA
| | | | | | - Eric A Weaver
- Department of Medicine, Division of Infectious Diseases, Mayo Clinic, Rochester, Minnesota, USA
| | - Larry R Pease
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| | - Richard J Webby
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Michael A Barry
- Department of Medicine, Division of Infectious Diseases, Mayo Clinic, Rochester, Minnesota, USA
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
13
|
Dai G, Rady HF, Huang W, Shellito JE, Mason C, Ramsay AJ. Gene-based neonatal immune priming potentiates a mucosal adenoviral vaccine encoding mycobacterial Ag85B. Vaccine 2016; 34:6267-6275. [PMID: 27823900 DOI: 10.1016/j.vaccine.2016.10.065] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 10/01/2016] [Accepted: 10/24/2016] [Indexed: 12/01/2022]
Abstract
Tuberculosis remains a major public health hazard worldwide, with neonates and young infants potentially more susceptible to infection than adults. BCG, the only vaccine currently available, provides some protection against tuberculous meningitis in children but variable efficacy in adults, and is not safe to use in immune compromised individuals. A safe and effective vaccine that could be given early in life, and that could also potentiate subsequent booster immunization, would represent a significant advance. To test this proposition, we have generated gene-based vaccine vectors expressing Ag85B from Mycobacterium tuberculosis (Mtb) and designed experiments to test their immunogenicity and protective efficacy particularly when given in heterologous prime-boost combination, with the initial DNA vaccine component given soon after birth. Intradermal delivery of DNA vaccines elicited Th1-based immune responses against Ag85B in neonatal mice but did not protect them from subsequent aerosol challenge with virulent Mtb H37Rv. Recombinant adenovirus vectors encoding Ag85B, given via the intranasal route at six weeks of age, generated moderate immune responses and were poorly protective. However, neonatal DNA priming following by mucosal boosting with recombinant adenovirus generated strong immune responses, as evidenced by strong Ag85B-specific CD4+ and CD8+ T cell responses, both in the lung-associated lymph nodes and the spleen, by the quality of these responding cells (assessed by their capacity to secrete multiple antimicrobial factors), and by improved protection, as indicated by reduced bacterial burden in the lungs following pulmonary TB challenge. These results suggest that neonatal immunization with gene-based vaccines may create a favorable immunological environment that potentiates the pulmonary mucosal boosting effects of a subsequent heterologous vector vaccine encoding the same antigen. Our data indicate that immunization early in life with mycobacterial antigens in an appropriate vaccine setting can prime for protective immunity against Mtb.
Collapse
Affiliation(s)
- Guixiang Dai
- Department of Microbiology, Immunology & Parasitology, LSUHSC-New Orleans, LA 70112, USA; The Louisiana Vaccine Center, LSUHSC-New Orleans, LA 70112, USA
| | - Hamada F Rady
- Department of Microbiology, Immunology & Parasitology, LSUHSC-New Orleans, LA 70112, USA; The Louisiana Vaccine Center, LSUHSC-New Orleans, LA 70112, USA
| | - Weitao Huang
- Department of Microbiology, Immunology & Parasitology, LSUHSC-New Orleans, LA 70112, USA; The Louisiana Vaccine Center, LSUHSC-New Orleans, LA 70112, USA
| | - Judd E Shellito
- Internal Medicine, LSUHSC-New Orleans, LA 70112, USA; The Louisiana Vaccine Center, LSUHSC-New Orleans, LA 70112, USA
| | - Carol Mason
- Internal Medicine, LSUHSC-New Orleans, LA 70112, USA
| | - Alistair J Ramsay
- Department of Microbiology, Immunology & Parasitology, LSUHSC-New Orleans, LA 70112, USA; Internal Medicine, LSUHSC-New Orleans, LA 70112, USA; The Louisiana Vaccine Center, LSUHSC-New Orleans, LA 70112, USA.
| |
Collapse
|
14
|
Singh S, Nehete PN, Yang G, He H, Nehete B, Hanley PW, Barry MA, Sastry KJ. Enhancement of Mucosal Immunogenicity of Viral Vectored Vaccines by the NKT Cell Agonist Alpha-Galactosylceramide as Adjuvant. Vaccines (Basel) 2015; 2:686-706. [PMID: 25553254 PMCID: PMC4278383 DOI: 10.3390/vaccines2040686] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Gene-based vaccination strategies, specifically viral vectors encoding vaccine immunogens are effective at priming strong immune responses. Mucosal routes offer practical advantages for vaccination by ease of needle-free administration, and immunogen delivery at readily accessible oral/nasal sites to efficiently induce immunity at distant gut and genital tissues. However, since mucosal tissues are inherently tolerant for induction of immune responses, incorporation of adjuvants for optimal mucosal vaccination strategies is important. We report here the effectiveness of alpha-galactosylceramide (α-GalCer), a synthetic glycolipid agonist of natural killer T (NKT) cells, as an adjuvant for enhancing immunogenicity of vaccine antigens delivered using viral vectors by mucosal routes in murine and nonhuman primate models. Significant improvement in adaptive immune responses in systemic and mucosal tissues was observed by including α-GalCer adjuvant for intranasal immunization of mice with vesicular stomatitis virus vector encoding the model antigen ovalbumin and adenoviral vectors expressing HIV env and Gag antigens. Activation of NKT cells in systemic and mucosal tissues along with significant increases in adaptive immune responses were observed in rhesus macaques immunized by intranasal and sublingual routes with protein or adenovirus vectored antigens when combined with α-GalCer adjuvant. These results support the utility of α-GalCer adjuvant for enhancing immunogenicity of mucosal vaccines delivered using viral vectors.
Collapse
Affiliation(s)
- Shailbala Singh
- Department of Immunology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; E-Mails: (S.S.); (G.Y.)
| | - Pramod N. Nehete
- Department of Veterinary Sciences, The University of Texas M.D. Anderson Cancer Center, Bastrop, TX 78602, USA; E-Mails: (P.N.N.); b (B.N.)
| | - Guojun Yang
- Department of Immunology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; E-Mails: (S.S.); (G.Y.)
| | - Hong He
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; E-Mail:
| | - Bharti Nehete
- Department of Veterinary Sciences, The University of Texas M.D. Anderson Cancer Center, Bastrop, TX 78602, USA; E-Mails: (P.N.N.); b (B.N.)
| | - Patrick W. Hanley
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health Rocky Mountain Laboratories, Hamilton, MT 59840, USA; E-Mail:
| | - Michael A. Barry
- Department of Internal Medicine, Division of Infectious Diseases, Mayo Clinic, Rochester, MN 55902, USA; E-Mail:
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55902, USA
- Department of Immunology, Mayo Clinic, Rochester, MN 55902, USA
- Translational Immunovirology and Biodefense Program, Mayo Clinic, Rochester, MN 55902, USA
| | - K. Jagannadha Sastry
- Department of Immunology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; E-Mails: (S.S.); (G.Y.)
- Department of Veterinary Sciences, The University of Texas M.D. Anderson Cancer Center, Bastrop, TX 78602, USA; E-Mails: (P.N.N.); b (B.N.)
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-713-563-3304; Fax: +1-713-563-3357
| |
Collapse
|
15
|
Ma J, Lu J, Huang H, Teng X, Tian M, Yu Q, Yuan X, Jing Y, Shi C, Li J, Fan X. Inhalation of recombinant adenovirus expressing granulysin protects mice infected with Mycobacterium tuberculosis. Gene Ther 2015; 22:968-76. [PMID: 26181627 DOI: 10.1038/gt.2015.73] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Revised: 06/22/2015] [Accepted: 07/08/2015] [Indexed: 11/09/2022]
Abstract
Granulysin is a cytolytic molecule with perforin and granzymes that is expressed by activated human CTLs, NK and γδ T cells, and it has broad antimicrobial activity, including to drug-sensitive and drug-resistant Mycobacterium tuberculosis. We hypothesized that approaches facilitating the expression of granulysin in M. tuberculosis-infected host cells in the lung may provide a novel treatment strategy for pulmonary TB. In this study, a recombinant replication-deficient adenovirus serotype 5-based rAdhGLi was constructed that expressed human granulysin in the cytosol of the U937 and RAW264.7 macrophage-like cell lines as confirmed by western blotting and co-localization technology using indirect immunofluorescence staining. Ninety-six hours after both cell lines were infected with M. tuberculosis, acid-fast staining and enumeration demonstrated that rAdhGLi-treated cells had a lower colony-forming units (CFU) of intracellular bacteria than culture medium or AdNull controls. Granulysin was only expressed in the lung and not in other organs following inhalation of rAdhGLi. In particular, immunocompetent BALB/c mice or SCID mice intranasally infected with ~200 CFU of virulent M. tuberculosis H37Rv intranasally were treated with rAdhGLi, and they showed decreased bacterial loads in the lung when compared with phosphate-buffered saline or AdNull controls. Importantly, a clear dose-dependent rAdhGLi treatment efficacy was found in infected BALB/c mice, with the most significant reduction in lung bacteria obtained in BALB/c mice treated with 10(9) plaque-forming units of rAdhGLi without any pathological changes. Our study indicates that rAdhGLi may be used as a novel and efficient treatment strategy with the capability to directly kill intracellular M. tuberculosis.
Collapse
Affiliation(s)
- J Ma
- Department of Pathogen Biology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, China
| | - J Lu
- Department of Pathogen Biology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, China
| | - H Huang
- Department of Pathogen Biology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, China
| | - X Teng
- Department of Pathogen Biology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, China
| | - M Tian
- Department of Pathogen Biology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, China
| | - Q Yu
- Department of Pathogen Biology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, China
| | - X Yuan
- Department of Pathogen Biology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, China
| | - Y Jing
- Department of Pathogen Biology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, China
| | - C Shi
- Department of Pathogen Biology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, China
| | - J Li
- Department of Pathogen Biology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, China
| | - X Fan
- Department of Pathogen Biology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
16
|
Sharma A, Wendland R, Sung B, Wu W, Grunwald T, Worgall S. Maternal immunization with chimpanzee adenovirus expressing RSV fusion protein protects against neonatal RSV pulmonary infection. Vaccine 2014; 32:5761-8. [PMID: 25171847 PMCID: PMC4713013 DOI: 10.1016/j.vaccine.2014.08.049] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2014] [Revised: 07/22/2014] [Accepted: 08/15/2014] [Indexed: 12/31/2022]
Abstract
Respiratory syncytial virus (RSV) is a leading cause of lower respiratory tract disease with high morbidity and mortality in young infants and children. Despite numerous efforts, a licensed vaccine against RSV remains elusive. Since young infants form the primary target group of RSV disease, maternal immunization to boost the protection in neonates is an attractive strategy. In this study we tested the efficacy of maternal immunization with a chimpanzee adenovirus expressing codon-optimized RSV fusion protein (AdC7-Fsyn) to protect infants against RSV infection. Single intranasal immunization of mice by AdC7-Fsyn induced robust anti-RSV systemic and mucosal immunity that protected against RSV without causing vaccine-enhanced RSV disease. RSV humoral immunity was transferred to pups born to immunized mothers that provided protection against RSV. Immunization with AdC7-Fsyn was effective even in the presence of Ad5 preimmunity. The maternally derived immunity was durable with the half-life of 14.63 days that reduced the viral replication up to 15 weeks of age. Notably, the passively immunized mice could be actively re-immunized with AdC7-Fsyn to boost and extend the protection. This substantiates maternal immunization with an AdC7-based vaccine expressing RSV F as feasible approach to protect against RSV early in life.
Collapse
Affiliation(s)
- Anurag Sharma
- Department of Genetic Medicine, Weill Cornell Medical College, New York, NY, United States
| | - Rebecca Wendland
- Department of Genetic Medicine, Weill Cornell Medical College, New York, NY, United States
| | - Biin Sung
- Department of Genetic Medicine, Weill Cornell Medical College, New York, NY, United States
| | - Wenzhu Wu
- Department of Genetic Medicine, Weill Cornell Medical College, New York, NY, United States
| | - Thomas Grunwald
- Department of Molecular and Medical Virology, Ruhr-University Bochum, Bochum, Germany
| | - Stefan Worgall
- Department of Genetic Medicine, Weill Cornell Medical College, New York, NY, United States; Department of Pediatrics, Weill Cornell Medical College, New York, NY, United States.
| |
Collapse
|
17
|
Differentially imprinted innate immunity by mucosal boost vaccination determines antituberculosis immune protective outcomes, independent of T-cell immunity. Mucosal Immunol 2013; 6:612-25. [PMID: 23131783 DOI: 10.1038/mi.2012.103] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Homologous and heterologous parenteral prime-mucosal boost immunizations have shown great promise in combating mucosal infections such as tuberculosis and AIDS. However, their immune mechanisms remain poorly defined. In particular, it is still unclear whether T-cell and innate immunity may be independently affected by these immunization modalities and how it impacts immune protective outcome. Using two virus-based tuberculosis vaccines (adenovirus (Ad) and vesicular stomatitis virus (VSV) vectors), we found that while both homologous (Ad/Ad) and heterologous (Ad/VSV) respiratory mucosal boost immunizations elicited similar T-cell responses in the lung, they led to drastically different immune protective outcomes. Compared with Ad-based boosting, VSV-based boosting resulted in poorly enhanced protection against tuberculosis. Such inferior protection was associated with differentially imprinted innate phagocytes, particularly the CD11c(+)CD11b(+/-) cells, in the lung. We identified heightened type 1 interferon (IFN) responses to be the triggering mechanism. Thus, increased IFN-β severely blunted interleukin-12 responses in infected phagocytes, which in turn impaired their nitric oxide production and antimycobacterial activities. Our study reveals that vaccine vectors may differentially imprint innate cells at the mucosal site of immunization, which can impact immune-protective outcome, independent of T-cell immunity, and it is of importance to determine both T-cell and innate cell immunity in vaccine studies.
Collapse
|
18
|
Alejo DM, Moraes MP, Liao X, Dias CC, Tulman ER, Diaz-San Segundo F, Rood D, Grubman MJ, Silbart LK. An adenovirus vectored mucosal adjuvant augments protection of mice immunized intranasally with an adenovirus-vectored foot-and-mouth disease virus subunit vaccine. Vaccine 2013; 31:2302-9. [PMID: 23499593 DOI: 10.1016/j.vaccine.2013.02.060] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Revised: 02/08/2013] [Accepted: 02/27/2013] [Indexed: 11/30/2022]
Abstract
Foot-and-mouth disease virus (FMDV) is a highly contagious pathogen that causes severe morbidity and economic losses to the livestock industry in many countries. The oral and respiratory mucosae are the main ports of entry of FMDV, so the stimulation of local immunity in these tissues may help prevent initial infection and viral spread. E. coli heat-labile enterotoxin (LT) has been described as one of the few molecules that have adjuvant activity at mucosal surfaces. The objective of this study was to evaluate the efficacy of replication-defective adenovirus 5 (Ad5) vectors encoding either of two LT-based mucosal adjuvants, LTB or LTR72. These vectored adjuvants were delivered intranasally to mice concurrent with an Ad5-FMDV vaccine (Ad5-A24) to assess their ability to augment mucosal and systemic humoral immune responses to Ad5-A24 and protection against FMDV. Mice receiving Ad5-A24 plus Ad5-LTR72 had higher levels of mucosal and systemic neutralizing antibodies than those receiving Ad5-A24 alone or Ad5-A24 plus Ad5-LTB. The vaccine plus Ad5-LTR72 group also demonstrated 100% survival after intradermal challenge with a lethal dose of homologous FMDV serotype A24. These results suggest that Ad5-LTR72 could be used as an important tool to enhance mucosal and systemic immunity against FMDV and potentially other pathogens with a common route of entry.
Collapse
Affiliation(s)
- Diana M Alejo
- Department of Animal Science, University of Connecticut, Storrs, CT 06269, United States
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
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: 58] [Impact Index Per Article: 4.8] [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
|
20
|
|
21
|
Hangalapura BN, Timares L, Oosterhoff D, Scheper RJ, Curiel DT, de Gruijl TD. CD40-targeted adenoviral cancer vaccines: the long and winding road to the clinic. J Gene Med 2012; 14:416-27. [PMID: 22228547 DOI: 10.1002/jgm.1648] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
The ability of dendritic cells (DCs) to orchestrate innate and adaptive immune responses has been exploited to develop potent anti-cancer immunotherapies. Recent clinical trials exploring the efficacy of ex vivo modified autologous DC-based vaccines have reported some promising results. However, in vitro generation of autologous DCs for clinical administration, their loading with tumor associated antigens (TAAs) and their activation, is laborious and expensive, and, as a result of inter-individual variability in the personalized vaccines, remains poorly standardized. An attractive alternative approach is to load resident DCs in vivo by targeted delivery of TAAs, using viral vectors and activating them simultaneously. To this end, we have constructed genetically-modified adenoviral (Ad) vectors and bispecific adaptor molecules to retarget Ad vectors encoding TAAs to the CD40 receptor on DCs. Pre-clinical human and murine studies conducted so far have clearly demonstrated the suitability of a 'two-component' (i.e. Ad and adaptor molecule) configuration for targeted modification of DCs in vivo for cancer immunotherapy. This review summarizes recent progress in the development of CD40-targeted Ad-based cancer vaccines and highlights pre-clinical issues in the clinical translation of this approach.
Collapse
Affiliation(s)
- Basav N Hangalapura
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
22
|
Wu Y, Wu S, Hou L, Wei W, Zhou M, Su Z, Wu J, Chen W, Ma G. Novel thermal-sensitive hydrogel enhances both humoral and cell-mediated immune responses by intranasal vaccine delivery. Eur J Pharm Biopharm 2012; 81:486-97. [PMID: 22507968 DOI: 10.1016/j.ejpb.2012.03.021] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2011] [Revised: 03/25/2012] [Accepted: 03/31/2012] [Indexed: 11/25/2022]
Abstract
A novel thermal sensitive hydrogel was formulated with N-[(2-hydroxy-3-trimethylammonium) propyl] chitosan chloride (HTCC) and α, β-glycerophosphate (α, β-GP). A serial of hydrogels containing different amount of GP and HTCC with diverse quarternize degree (QD, 41%, 59%, 79.5%, and 99%) were prepared and characterized by rheological method. The hydrogel was subsequently evaluated for intranasal vaccine delivery with adenovirus based Zaire Ebola virus glycoprotein antigen (Ad-GPZ). Results showed that moderate quarternized HTCC (60% and 79.5%) hydrogel/antigen formulations induced highest IgG, IgG1, and IgG2a antibody titers in serum, as well as mucosal IgA responses in lung wash, which may attributed to the prolonged antigen residence time due to the thermal-sensitivity of this hydrogel. Furthermore, CD8(+) splenocytes for IFN-γ positive cell assay and the release profile of Th1/Th2 type cytokines (IFN-γ, IL-2, IL-10, and IL-4) showed that hydrogel/Ad-GPZ generated an overwhelmingly enhanced Th1 biased cellular immune response. In addition, this hydrogel displayed low toxicity to nasal tissue and epithelial cells even by frequently intranasal dosing of hydrogel. All these results strongly supported this hydrogel as a safe and effective delivery system for nasal immunization.
Collapse
Affiliation(s)
- Youbin Wu
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
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: 32] [Impact Index Per Article: 2.3] [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
|
24
|
Moraes TJ, Lin GH, Wen T, Watts TH. Incorporation of 4-1BB ligand into an adenovirus vaccine vector increases the number of functional antigen-specific CD8 T cells and enhances the duration of protection against influenza-induced respiratory disease. Vaccine 2011; 29:6301-12. [DOI: 10.1016/j.vaccine.2011.06.022] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2010] [Revised: 04/07/2011] [Accepted: 06/04/2011] [Indexed: 11/30/2022]
|
25
|
Hangalapura BN, Oosterhoff D, de Groot J, Boon L, Tüting T, van den Eertwegh AJ, Gerritsen WR, van Beusechem VW, Pereboev A, Curiel DT, Scheper RJ, de Gruijl TD. Potent antitumor immunity generated by a CD40-targeted adenoviral vaccine. Cancer Res 2011; 71:5827-37. [PMID: 21747119 DOI: 10.1158/0008-5472.can-11-0804] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In situ delivery of tumor-associated antigen (TAA) genes into dendritic cells (DC) has great potential as a generally applicable tumor vaccination approach. Although adenoviruses (Ad) are an attractive vaccine vehicle in this regard, Ad-mediated transduction of DCs is hampered by the lack of expression of the Ad receptor CAR on the DC surface. DC activation also requires interaction of CD40 with its ligand CD40L to generate protective T-cell-mediated tumor immunity. Therefore, to create a strategy to target Ads to DCs in vivo, we constructed a bispecific adaptor molecule with the CAR ectodomain linked to the CD40L extracellular domain via a trimerization motif (CFm40L). By targeting Ad to CD40 with the use of CFm40L, we enhanced both transduction and maturation of cultured bone marrow-derived DCs. Moreover, we improved transduction efficiency of DCs in lymph node and splenic cell suspensions in vitro and in skin and vaccination site-draining lymph nodes in vivo. Furthermore, CD40 targeting improved the induction of specific CD8(+) T cells along with therapeutic efficacy in a mouse model of melanoma. Taken together, our findings support the use of CD40-targeted Ad vectors encoding full-length TAA for in vivo targeting of DCs and high-efficacy induction of antitumor immunity.
Collapse
Affiliation(s)
- Basav N Hangalapura
- Department of Medical Oncology and Pathology, VU University Medical Center, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Tutykhina IL, Logunov DY, Shcherbinin DN, Shmarov MM, Tukhvatulin AI, Naroditsky BS, Gintsburg AL. Development of adenoviral vector-based mucosal vaccine against influenza. J Mol Med (Berl) 2011; 89:331-41. [PMID: 21104066 DOI: 10.1007/s00109-010-0696-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2010] [Revised: 09/24/2010] [Accepted: 10/13/2010] [Indexed: 12/25/2022]
Abstract
The recent pandemic threat of the influenza virus makes the increased safety and efficiency of vaccination against the pathogen a most important issue. It has been well established that for maximum protective effect, the vaccination should mimic natural infection. Therefore, recent efforts to develop a new influenza vaccine have focused on intranasal immunization strategies. Intranasal immunization is capable of inducing secretory IgA and serum IgG responses to provide a double defense against mucosal pathogens. On the other hand, it is desirable that a live pathogen is not present in the vaccine. In addition, for optimal induction of the immune responses via the nasal route, efficient and safe mucosal adjuvants are also required. This is possible to attain using an adenoviral vector for vaccine development. Adenoviral vectors are capable of delivering and protecting the antigen encoding sequence. They also possess a natural mechanism for penetrating into the nasal mucous membrane and are capable of activating the innate immune response. This review describes the basic prerequisites for the involvement of recombinant adenoviruses for mucosal (nasal) vaccine development against the influenza virus.
Collapse
Affiliation(s)
- Irina L Tutykhina
- Laboratory of Molecular Biotechnology, Gamaleya Research Institute of Epidemiology and Microbiology, ul. Gamaleya 18, Moscow 123098, Russia
| | | | | | | | | | | | | |
Collapse
|
27
|
Hangalapura BN, Oosterhoff D, Gupta T, de Groot J, Wijnands PGJTB, van Beusechem VW, den Haan J, Tüting T, van den Eertwegh AJM, Curiel DT, Scheper RJ, de Gruijl TD. Delivery route, MyD88 signaling and cross-priming events determine the anti-tumor efficacy of an adenovirus based melanoma vaccine. Vaccine 2011; 29:2313-21. [PMID: 21272606 PMCID: PMC3058128 DOI: 10.1016/j.vaccine.2011.01.022] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2010] [Revised: 12/30/2010] [Accepted: 01/11/2011] [Indexed: 02/06/2023]
Abstract
Adenovirus (Ad)-based vaccines are considered for cancer immunotherapy, yet, detailed knowledge on their mechanism of action and optimal delivery route for anti-tumor efficacy is lacking. Here, we compared the anti-tumor efficacy of an Ad-based melanoma vaccine after intradermal, intravenous, intranasal or intraperitoneal delivery in the B16F10 melanoma model. The intradermal route induced superior systemic anti-melanoma immunity which was MyD88 signaling-dependent. Predominant transduction of non-professional antigen-presenting cells at the dermal vaccination sites and draining lymph nodes, suggested a role for cross-presentation, which was confirmed in vitro. We conclude that the dermis provides an optimal route of entry for Ad-based vaccines for high-efficacy systemic anti-tumor immunization and that this immunization likely involves cross-priming events in the draining lymph nodes.
Collapse
Affiliation(s)
- Basav N Hangalapura
- Dept of Medical Oncology, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Dinja Oosterhoff
- Dept of Medical Oncology, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Tarun Gupta
- Dept of Medical Oncology, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Jan de Groot
- Dept of Pathology, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Pepijn GJTB Wijnands
- Dept of Medical Oncology, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Dept of Pathology, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | | | - Joke den Haan
- Dept of Cell Biology and Immunology of the VU University medical center and Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Thomas Tüting
- Dept of Dermatology, Lab of Experimental Dermatology, University of Bonn. Bonn, Germany
| | | | - David T Curiel
- Division of Human Gene Therapy, University of Alabama at Birmingham, Birmingham AL
| | - Rik J Scheper
- Dept of Pathology, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Tanja D de Gruijl
- Dept of Medical Oncology, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
28
|
Abstract
Tuberculosis in infants and young children remains an all too common cause of morbidity and mortality in high burden countries, despite the fact that the majority of these children receive vaccination with BCG in infancy. BCG confers incomplete and variable protection against pulmonary tuberculosis [PTB] and is unsafe in HIV positive persons. Newer TB vaccines, which, it is hoped, will either replace or complement BCG are being developed and a number of these have reached the stage of clinical trials, with two booster vaccines going into Phase IIB trials in 2009. Prospects for at least one new licensed TB vaccine within the next 5-10 years appear reasonable. This article explores some of the issues around the development of new vaccines against TB and details the leading candidates.
Collapse
Affiliation(s)
- Tony Hawkridge
- Aeras Global TB Vaccine Foundation, Africa Office, Belmont Square, Belmont Road, Rondebosch, Cape Town, 7700, South Africa.
| | | |
Collapse
|
29
|
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
|
30
|
Swayze RD, Bhogal HS, Barabé ND, McLaws LJ, Wu JQH. Envelope protein E1 as vaccine target for western equine encephalitis virus. Vaccine 2010; 29:813-20. [PMID: 21084062 DOI: 10.1016/j.vaccine.2010.11.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2010] [Revised: 10/29/2010] [Accepted: 11/02/2010] [Indexed: 10/18/2022]
Abstract
Western equine encephalitis virus (WEEV) is a mosquito-borne RNA virus which causes lethal infection in humans and equines. There are no commercial vaccines or anti-WEEV drugs available for humans. We used replication-defective, human adenovirus serotype-5 (HAd5) as a delivery vector for developing WEEV vaccine. Our previous study found delivery of both E1 and E2 envelope proteins of WEEV by HAd5 vector offers complete protection against lethal challenge of WEEV. In this paper, we constructed a HAd5-vectored E1 vaccine, Ad5-E1. Mice given single-dose vaccination of Ad5-E1 were completely protected against both homologous and heterologous WEEV strains. The protection was rapid, which was achieved as early as day 7 after vaccination. In addition, Ad5-E1 induced a strong WEEV-specific T cell response. Our data suggest E1 is a potential target for developing single-dose, fast-acting, HAd5-vectored vaccine for WEEV.
Collapse
Affiliation(s)
- Richard D Swayze
- Defence Research and Development Canada - Suffield, Box 4000, Station Main, Medicine Hat, Alberta T1A 8K6, Canada
| | | | | | | | | |
Collapse
|
31
|
Zhang H, Wen K, Shen J, Geng S, Huang J, Pan Z, Jiao X. Characterization of immune responses following intranasal immunization with the Mycobacterium bovis CFP-10 protein expressed by attenuated Salmonella typhimurium. Scand J Immunol 2010; 72:277-83. [PMID: 20883312 DOI: 10.1111/j.1365-3083.2010.02421.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Culture filtrate protein 10 (CFP-10) from Mycobacterium bovis or Mycobacterium tuberculosis (MTB) is an immunodominant T-cell antigen expressed during the early stages of infection. Because lungs are most commonly associated with primary M. bovis infections, specific immunity at this site is desirable for protection. Therefore, in this study, immune responses generated in mouse lung, spleen and Peyer's patches were examined following intranasal (i.n.) immunization with Salmonella typhimurium- expressing CFP-10. Cells harvested from the lungs and Peyer's patches of immunized mice and then stimulated with CFP-10 produced significant levels of IFN-γ and these mice developed elevated serum IgG and lung IgA anti-CFP-10 responses, suggesting that this approach induced potent anti-CFP-10 mucosal immunity. Our study demonstrates that i.n. administration of CFP-10 expressed by S. typhimurium represents an effective way to induce efficient immune response to M. bovis antigen.
Collapse
Affiliation(s)
- H Zhang
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, Jiangsu, China
| | | | | | | | | | | | | |
Collapse
|
32
|
Assessment of route of administration and dose escalation for an adenovirus-based influenza A Virus (H5N1) vaccine in chickens. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2010; 17:1467-72. [PMID: 20660133 DOI: 10.1128/cvi.00180-10] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Highly pathogenic avian influenza (HPAI) virus causes one of the most economically devastating poultry diseases. An HPAI vaccine to prevent the disease in commercial and backyard birds must be effective, safe, and inexpensive. Recently, we demonstrated the efficacy of an adenovirus-based H5N1 HPAI vaccine (Ad5.HA) in chickens. To further evaluate the potential of the Ad5.HA vaccine and its cost-effectiveness, studies to determine the minimal effective dose and optimal route of administration in chickens were performed. A dose as low as 10(7) viral particles (vp) of adenovirus-based H5N1 vaccine per chicken was sufficient to generate a robust humoral immune response, which correlated with the previously reported level of protection. Several routes of administration, including intratracheal, conjunctival, subcutaneous, and in ovo routes, were evaluated for optimal vaccine administration. However, only the subcutaneous route of immunization induced a satisfactory level of influenza virus-specific antibodies. Importantly, these studies established that the vaccine-induced immunity was cross-reactive against an H5N1 strain from a different clade, emphasizing the potential of cross-protection. Our results suggest that the Ad5.HA HPAI vaccine is safe and effective, with the potential of cross-clade protection. The ease of manufacturing and cost-effectiveness make Ad5.HA an excellent avian influenza vaccine candidate with the ability to protect poultry from HPAI virus infection. Considering the limitations of the influenza vaccine technology currently used for poultry applications, any effort aimed at overcoming those limitations is highly significant.
Collapse
|
33
|
Clapp B, Golden S, Maddaloni M, Staats HF, Pascual DW. Adenovirus F protein as a delivery vehicle for botulinum B. BMC Immunol 2010; 11:36. [PMID: 20609248 PMCID: PMC2912244 DOI: 10.1186/1471-2172-11-36] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2010] [Accepted: 07/07/2010] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Immunization with recombinant carboxyl-terminal domain of the heavy chain (Hc domain) of botulinum neurotoxin (BoNT) stimulates protective immunity against native BoNT challenge. Most studies developing a botulism vaccine have focused on the whole Hc; however, since the principal protective epitopes are located within beta-trefoil domain (Hcbetatre), we hypothesize that immunization with the Hcbetatre domain is sufficient to confer protective immunity. In addition, enhancing its uptake subsequent to nasal delivery prompted development of an alternative vaccine strategy, and we hypothesize that the addition of targeting moiety adenovirus 2 fiber protein (Ad2F) may enhance such uptake during vaccination. RESULTS The Hcbetatre serotype B immunogen was genetically fused to Ad2F (Hcbetatre/B-Ad2F), and its immunogenicity was tested in mice. In combination with the mucosal adjuvant, cholera toxin (CT), enhanced mucosal IgA and serum IgG Ab titers were induced by nasal Hcbetatre-Ad2F relative to Hcbetatre alone; however, similar Ab titers were obtained upon intramuscular immunization. These BoNT/B-specific Abs induced by nasal immunization were generally supported in large part by Th2 cells, as opposed to Hcbetatre-immunized mice that showed more mixed Th1 and Th2 cells. Using a mouse neutralization assay, sera from animals immunized with Hcbetatre and Hcbetatre-Ad2F protected mice against 2.0 LD50. CONCLUSION These results demonstrate that Hcbetatre-based immunogens are highly immunogenic, especially when genetically fused to Ad2F, and Ad2F can be exploited as a vaccine delivery platform to the mucosa.
Collapse
Affiliation(s)
- Beata Clapp
- Veterinary Molecular Biology, Montana State University, Bozeman, MT 59717-3610, USA
| | - Sarah Golden
- Veterinary Molecular Biology, Montana State University, Bozeman, MT 59717-3610, USA
| | - Massimo Maddaloni
- Veterinary Molecular Biology, Montana State University, Bozeman, MT 59717-3610, USA
| | - Herman F Staats
- Department of Pathology, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - David W Pascual
- Veterinary Molecular Biology, Montana State University, Bozeman, MT 59717-3610, USA
| |
Collapse
|
34
|
Kohlmann R, Schwannecke S, Tippler B, Ternette N, Temchura VV, Tenbusch M, Überla K, Grunwald T. Protective efficacy and immunogenicity of an adenoviral vector vaccine encoding the codon-optimized F protein of respiratory syncytial virus. J Virol 2009; 83:12601-10. [PMID: 19776123 PMCID: PMC2786764 DOI: 10.1128/jvi.01036-09] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2009] [Accepted: 09/17/2009] [Indexed: 11/20/2022] Open
Abstract
Adenoviral vectors (AdV) have received considerable attention for vaccine development because of their high immunogenicity and efficacy. In previous studies, it was shown that DNA immunization of mice with codon-optimized expression plasmids encoding the fusion protein of respiratory syncytial virus (RSV F) resulted in enhanced protection against RSV challenge compared to immunization with plasmids carrying the wild-type cDNA sequence of RSV F. In this study, we constructed AdV carrying the codon-optimized full-length RSV F gene (AdV-F) or the soluble form of the RSV F gene (AdV-Fsol). BALB/c mice were immunized twice with AdV-F or AdV-Fsol and challenged with RSV intranasally. Substantial levels of antibody to RSV F were induced by both AdV vaccines, with peak neutralizing-antibody titers of 1:900. Consistently, the viral loads in lung homogenates and bronchoalveolar lavage fluids were significantly reduced by a factor of more than 60,000. The protection against viral challenge could be measured even 8 months after the booster immunization. AdV-F and AdV-Fsol induced similar levels of immunogenicity and protective efficacy. Therefore, these results encourage further development of AdV vaccines against RSV infection in humans.
Collapse
Affiliation(s)
- Rebekka Kohlmann
- Department of Molecular and Medical Virology, Ruhr-Universitaet Bochum, Bochum, Germany
| | - Sarah Schwannecke
- Department of Molecular and Medical Virology, Ruhr-Universitaet Bochum, Bochum, Germany
| | - Bettina Tippler
- Department of Molecular and Medical Virology, Ruhr-Universitaet Bochum, Bochum, Germany
| | - Nicola Ternette
- Department of Molecular and Medical Virology, Ruhr-Universitaet Bochum, Bochum, Germany
| | - Vladimir V. Temchura
- Department of Molecular and Medical Virology, Ruhr-Universitaet Bochum, Bochum, Germany
| | - Matthias Tenbusch
- Department of Molecular and Medical Virology, Ruhr-Universitaet Bochum, Bochum, Germany
| | - Klaus Überla
- Department of Molecular and Medical Virology, Ruhr-Universitaet Bochum, Bochum, Germany
| | - Thomas Grunwald
- Department of Molecular and Medical Virology, Ruhr-Universitaet Bochum, Bochum, Germany
| |
Collapse
|
35
|
Hoegh-Petersen M, Thomsen AR, Christensen JP, Holst PJ. Mucosal immunization with recombinant adenoviral vectors expressing murine gammaherpesvirus-68 genes M2 and M3 can reduce latent viral load. Vaccine 2009; 27:6723-30. [DOI: 10.1016/j.vaccine.2009.08.104] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2008] [Revised: 07/13/2009] [Accepted: 08/26/2009] [Indexed: 12/22/2022]
|
36
|
Intranasal mucosal boosting with an adenovirus-vectored vaccine markedly enhances the protection of BCG-primed guinea pigs against pulmonary tuberculosis. PLoS One 2009; 4:e5856. [PMID: 19516906 PMCID: PMC2689939 DOI: 10.1371/journal.pone.0005856] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2009] [Accepted: 05/15/2009] [Indexed: 11/19/2022] Open
Abstract
Background Recombinant adenovirus-vectored (Ad) tuberculosis (TB) vaccine platform has demonstrated great potential to be used either as a stand-alone or a boost vaccine in murine models. However, Ad TB vaccine remains to be evaluated in a more relevant and sensitive guinea pig model of pulmonary TB. Many vaccine candidates shown to be effective in murine models have subsequently failed to pass the test in guinea pig models. Methods and Findings Specific pathogen-free guinea pigs were immunized with BCG, AdAg85A intranasally (i.n), AdAg85A intramuscularly (i.m), BCG boosted with AdAg85A i.n, BCG boosted with AdAg85A i.m, or treated only with saline. The animals were then infected by a low-dose aerosol of M. tuberculosis (M.tb). At the specified times, the animals were sacrificed and the levels of infection in the lung and spleen were assessed. In separate studies, the long-term disease outcome of infected animals was monitored until the termination of this study. Immunization with Ad vaccine alone had minimal beneficial effects. Immunization with BCG alone and BCG prime-Ad vaccine boost regimens significantly reduced the level of M.tb infection in the tissues to a similar extent. However, while BCG alone prolonged the survival of infected guinea pigs, the majority of BCG-immunized animals succumbed by 53 weeks post-M.tb challenge. In contrast, intranasal or intramuscular Ad vaccine boosting of BCG-primed animals markedly improved the survival rate with 60% of BCG/Ad i.n- and 40% of BCG/Ad i.m-immunized guinea pigs still surviving by 74 weeks post-aerosol challenge. Conclusions Boosting, particularly via the intranasal mucosal route, with AdAg85A vaccine is able to significantly enhance the long-term survival of BCG-primed guinea pigs following pulmonary M.tb challenge. Our results thus support further evaluation of this viral-vectored TB vaccine in clinical trials.
Collapse
|
37
|
Antigen delivery systems for veterinary vaccine development. Viral-vector based delivery systems. Vaccine 2009; 26:6508-28. [PMID: 18838097 PMCID: PMC7131726 DOI: 10.1016/j.vaccine.2008.09.044] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2008] [Revised: 08/21/2008] [Accepted: 09/16/2008] [Indexed: 11/30/2022]
Abstract
The recent advances in molecular genetics, pathogenesis and immunology have provided an optimal framework for developing novel approaches in the rational design of vaccines effective against viral epizootic diseases. This paper reviews most of the viral-vector based antigen delivery systems (ADSs) recently developed for vaccine testing in veterinary species, including attenuated virus and DNA and RNA viral vectors. Besides their usefulness in vaccinology, these ADSs constitute invaluable tools to researchers for understanding the nature of protective responses in different species, opening the possibility of modulating or potentiating relevant immune mechanisms involved in protection.
Collapse
|
38
|
Immunization with a bivalent adenovirus-vectored tuberculosis vaccine provides markedly improved protection over its monovalent counterpart against pulmonary tuberculosis. Mol Ther 2009; 17:1093-100. [PMID: 19319120 DOI: 10.1038/mt.2009.60] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Recombinant virus-vectored vaccines hold great promise for tuberculosis (TB) vaccination strategies. However, there is a lack of side-by-side comparative investigations to dissect the functional differences and support the advantage of multivalent virus-vectored vaccine over its monovalent counterpart. We previously successfully developed a monovalent adenovirus (Ad)-vectored vaccine expressing Ag85a (AdAg85a) and demonstrated its superior protective efficacy in models of pulmonary TB. In this study, we have developed a bivalent Ad TB vaccine expressing Ag85a and TB10.4 antigens as a fusion protein (AdAg85a:TB10.4) and compared its T-cell-activating and immune protective efficacy with that by monovalent AdAg85a. A single intranasal (i.n.) administration of AdAg85a:TB10.4 induced robust T-cell responses toward the respective antigens within the airway lumen and spleen, although the level of Ag85a-specific T-cell responses in the airway lumen triggered by bivalent AdAg85a:TB10.4 was lower than that by its monovalent counterpart at earlier time points. Thus, a single i.n. delivery of AdAg85a:TB10.4 conferred a markedly improved and sustained level of protection in the lung against Mycobacterium tuberculosis (M.tb) challenge over that by AdAg85a or by conventional BCG immunization with similarly induced levels of protection in the spleen. Our results indicate a unique advantage of multivalent viral-vectored TB vaccines for immunization against pulmonary TB.
Collapse
|
39
|
Zhang Q, Su X, Seto D, Zheng BJ, Tian X, Sheng H, Li H, Wang Y, Zhou R. Construction and characterization of a replication-competent human adenovirus type 3-based vector as a live-vaccine candidate and a viral delivery vector. Vaccine 2009; 27:1145-53. [DOI: 10.1016/j.vaccine.2008.12.039] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2008] [Revised: 12/07/2008] [Accepted: 12/20/2008] [Indexed: 01/02/2023]
|
40
|
Gag-specific immune enhancement of lentiviral infection after vaccination with an adenoviral vector in an animal model of AIDS. Vaccine 2009; 27:928-39. [DOI: 10.1016/j.vaccine.2008.11.086] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2008] [Revised: 10/27/2008] [Accepted: 11/06/2008] [Indexed: 12/22/2022]
|
41
|
Demberg T, Robert-Guroff M. Mucosal immunity and protection against HIV/SIV infection: strategies and challenges for vaccine design. Int Rev Immunol 2009; 28:20-48. [PMID: 19241252 PMCID: PMC3466469 DOI: 10.1080/08830180802684331] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
To date, most HIV vaccine strategies have focused on parenteral immunization and systemic immunity. These approaches have not yielded the efficacious HIV vaccine urgently needed to control the AIDS pandemic. As HIV is primarily mucosally transmitted, efforts are being re-focused on mucosal vaccine strategies, in spite of complexities of immune response induction and evaluation. Here, we outline issues in mucosal vaccine design and illustrate strategies with examples from the recent literature. Development of a successful HIV vaccine will require in-depth understanding of the mucosal immune system, knowledge that ultimately will benefit vaccine design for all mucosally transmitted infectious agents.
Collapse
Affiliation(s)
- Thorsten Demberg
- Vaccine Branch, National Cancer Institute, NIH, Bethesda, MD 20892
| | | |
Collapse
|
42
|
|
43
|
Croyle MA, Patel A, Tran KN, Gray M, Zhang Y, Strong JE, Feldmann H, Kobinger GP. Nasal delivery of an adenovirus-based vaccine bypasses pre-existing immunity to the vaccine carrier and improves the immune response in mice. PLoS One 2008; 3:e3548. [PMID: 18958172 PMCID: PMC2569416 DOI: 10.1371/journal.pone.0003548] [Citation(s) in RCA: 146] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2008] [Accepted: 09/28/2008] [Indexed: 11/30/2022] Open
Abstract
Pre-existing immunity to human adenovirus serotype 5 (Ad5) is common in the general population. Bypassing pre-existing immunity could maximize Ad5 vaccine efficacy. Vaccination by the intramuscular (I.M.), nasal (I.N.) or oral (P.O.) route with Ad5 expressing Ebola Zaire glycoprotein (Ad5-ZGP) fully protected naïve mice against lethal challenge with Ebola. In the presence of pre-existing immunity, only mice vaccinated I.N. survived. The frequency of IFN-γ+ CD8+ T cells was reduced by 80% and by 15% in animals vaccinated by the I.M. and P.O. routes respectively. Neutralizing antibodies could not be detected in serum from either treatment group. Pre-existing immunity did not compromise the frequency of IFN-γ+ CD8+ T cells (3.9±1% naïve vs. 3.6±1% pre-existing immunity, PEI) nor anti-Ebola neutralizing antibody (NAB, 40±10 reciprocal dilution, both groups). The number of INF-γ+ CD8+ cells detected in bronchioalveolar lavage fluid (BAL) after I.N. immunization was not compromised by pre-existing immunity to Ad5 (146±14, naïve vs. 120±16 SFC/million MNCs, PEI). However, pre-existing immunity reduced NAB levels in BAL by ∼25% in this group. To improve the immune response after oral vaccination, the Ad5-based vaccine was PEGylated. Mice given the modified vaccine did not survive challenge and had reduced levels of IFN-γ+ CD8+ T cells 10 days after administration (0.3±0.3% PEG vs. 1.7±0.5% unmodified). PEGylation did increase NAB levels 2-fold. These results provide some insight about the degree of T and B cell mediated immunity necessary for protection against Ebola virus and suggest that modification of the virus capsid can influence the type of immune response elicited by an Ad5-based vaccine.
Collapse
Affiliation(s)
- Maria A. Croyle
- Division of Pharmaceutics, College of Pharmacy, The University of Texas at Austin, Austin, Texas, United States of America
- Institute of Cellular and Molecular Biology, The University of Texas at Austin, Austin, Texas, United States of America
| | - Ami Patel
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada
- Department of Medical Microbiology, University of Manitoba, Winnipeg, Canada
| | - Kaylie N. Tran
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada
| | - Michael Gray
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada
| | - Yi Zhang
- Department of Internal Medicine, Division of Immunology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - James E. Strong
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada
- Department of Medical Microbiology, University of Manitoba, Winnipeg, Canada
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, Canada
| | - Heinz Feldmann
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada
- Department of Medical Microbiology, University of Manitoba, Winnipeg, Canada
| | - Gary P. Kobinger
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada
- Department of Medical Microbiology, University of Manitoba, Winnipeg, Canada
- * E-mail:
| |
Collapse
|
44
|
Jeyanathan M, Mu J, Kugathasan K, Zhang X, Damjanovic D, Small C, Divangahi M, Petrof BJ, Hogaboam CM, Xing Z. Airway Delivery of Soluble Mycobacterial Antigens Restores Protective Mucosal Immunity by Single Intramuscular Plasmid DNA Tuberculosis Vaccination: Role of Proinflammatory Signals in the Lung. THE JOURNAL OF IMMUNOLOGY 2008; 181:5618-26. [DOI: 10.4049/jimmunol.181.8.5618] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
45
|
Peek LJ, Middaugh CR, Berkland C. Nanotechnology in vaccine delivery. Adv Drug Deliv Rev 2008; 60:915-28. [PMID: 18325628 PMCID: PMC7103321 DOI: 10.1016/j.addr.2007.05.017] [Citation(s) in RCA: 371] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2007] [Accepted: 05/01/2007] [Indexed: 01/19/2023]
Abstract
With very few adjuvants currently being used in marketed human vaccines, a critical need exists for novel immunopotentiators and delivery vehicles capable of eliciting humoral, cellular and mucosal immunity. Such crucial vaccine components could facilitate the development of novel vaccines for viral and parasitic infections, such as hepatitis, HIV, malaria, cancer, etc. In this review, we discuss clinical trial results for various vaccine adjuvants and delivery vehicles being developed that are approximately nanoscale (< 1000 nm) in size. Humoral immune responses have been observed for most adjuvants and delivery platforms while only viral vectors, ISCOMs and Montanide™ ISA 51 and 720 have shown cytotoxic T cell responses in the clinic. MF59 and MPL® have elicited Th1 responses, and virus-like particles, non-degradable nanoparticles and liposomes have also generated cellular immunity. Such vaccine components have also been evaluated for alternative routes of administration with clinical successes reported for intranasal delivery of viral vectors and proteosomes and oral delivery of a VLP vaccine.
Collapse
|
46
|
Impact of recombinant adenovirus serotype 35 priming versus boosting of a Plasmodium falciparum protein: characterization of T- and B-cell responses to liver-stage antigen 1. Infect Immun 2008; 76:1709-18. [PMID: 18212075 DOI: 10.1128/iai.01614-07] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Prime-boost vaccination regimens with heterologous antigen delivery systems have indicated that redirection of the immune response is feasible. We showed earlier that T-cell responses to circumsporozoite (CS) protein improved significantly when the protein is primed with recombinant adenovirus serotype 35 coding for CS (rAd35.CS). The current study was designed to answer the question whether such an effect can be extended to liver-stage antigens (LSA) of Plasmodium falciparum such as LSA-1. Studies with mice have demonstrated that the LSA-1 protein induces strong antibody response but a weak T-cell immunity. We first identified T-cell epitopes in LSA-1 by use of intracellular gamma interferon (IFN-gamma) staining and confirmed these epitopes by means of enzyme-linked immunospot assay and pentamer staining. We show that a single immunization with rAd35.LSA-1 induced a strong antigen-specific IFN-gamma CD8(+) T-cell response but no measurable antibody response. In contrast, vaccinations with the adjuvanted recombinant LSA-1 protein induced remarkably low cellular responses but strong antibody responses. Finally, both priming and boosting of the adjuvanted protein by rAd35 resulted in enhanced T-cell responses without impairing the level of antibody responses induced by the protein immunizations alone. Furthermore, the incorporation of rAd35 in the vaccination schedule led to a skewing of LSA-1-specific antibody responses toward a Th1-type immune response. Our results show the ability of rAd35 to induce potent T-cell immunity in combination with protein in a prime-boost schedule without impairing the B-cell response.
Collapse
|
47
|
Huang D, Pereboev AV, Korokhov N, He R, Larocque L, Gravel C, Jaentschke B, Tocchi M, Casley WL, Lemieux M, Curiel DT, Chen W, Li X. Significant alterations of biodistribution and immune responses in Balb/c mice administered with adenovirus targeted to CD40(+) cells. Gene Ther 2007; 15:298-308. [PMID: 18046426 PMCID: PMC7091597 DOI: 10.1038/sj.gt.3303085] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
CD40 ligation has been shown to promote antigen-presenting functions of dendritic cells, which express CD40 receptor. Here we reported significantly altered biodistribution and immune responses with the use of CD40-targeted adenovirus. Compared with unmodified adenovirus 5, the CD40-targeted adenovirus following intravenous administration (i.v.) resulted in increased transgene expressions in the lung and thymus, which normally do not take up significant amounts of adenovirus. Intradermal injection saw modified adenovirus being mainly processed in local draining lymph nodes and skin. Following intranasal administration (i.n.), neither unmodified nor targeted viruses were found to be in the liver or spleen, which predominantly took up the virus following i.v. administration. However, inadvertent infection of the brain was found with unmodified adenoviruses, with the second highest gene expression among 14 tissues examined. Importantly, such undesirable effects were largely ablated with the use of targeted vector. Moreover, the targeted adenovirus elicited more sustained antigen-specific cellular immune responses (up to 17-fold) at later time points (30 days post boosting), but also significantly hampered humoral responses irrespective of administration routes. Additional data suggest the skewed immune responses induced by the targeted adenoviruses were not due to the identity of the transgene but more likely a combination of overall transgene load and CD40 stimulation.
Collapse
Affiliation(s)
- D Huang
- Centre for Biologics Research, Biologics and Genetic Therapies Directorate, HPFB, Health Canada, Ottawa, Ontario, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Patel A, Zhang Y, Croyle M, Tran K, Gray M, Strong J, Feldmann H, Wilson J, Kobinger G. Mucosal Delivery of Adenovirus‐Based Vaccine Protects against Ebola Virus Infection in Mice. J Infect Dis 2007; 196 Suppl 2:S413-20. [DOI: 10.1086/520603] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
49
|
Xing Z, Charters TJ, Carters TJ. Heterologous boost vaccines for bacillus Calmette-Guérin prime immunization against tuberculosis. Expert Rev Vaccines 2007; 6:539-46. [PMID: 17669008 DOI: 10.1586/14760584.6.4.539] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The current tuberculosis (TB) epidemic continues to call for the development of effective vaccination strategies. The initial TB vaccine research effort mostly focused on the search for a vaccine that might be better than, and thus could replace, the current bacillus Calmette-Guérin (BCG) vaccine. It has increasingly been realized that BCG or an improved BCG will continue to be used as a prime TB vaccine and there is a need to develop effective boost vaccines that could enhance and prolong the protective immunity of BCG prime immunization. Mounting experimental evidence suggests that recombinant vaccines, including both recombinant protein and genetic vector vaccines, are effective in boosting immune activation and protection by BCG vaccination. This review will discuss recent advances and the authors' views in the development of there boost vaccines.
Collapse
Affiliation(s)
- Zhou Xing
- McMaster University, Infectious Diseases Division, Centre for Gene Therapeutics and Department of Pathology and Molecular Medicine, Hamilton, Ontario, L8N 3Z5, Canada.
| | | | | |
Collapse
|
50
|
Brandtzaeg P. Induction of secretory immunity and memory at mucosal surfaces. Vaccine 2007; 25:5467-84. [PMID: 17227687 DOI: 10.1016/j.vaccine.2006.12.001] [Citation(s) in RCA: 347] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2006] [Revised: 11/08/2006] [Accepted: 12/01/2006] [Indexed: 11/27/2022]
Abstract
Mucosal epithelia comprise an extensive vulnerable barrier which is reinforced by numerous innate defence mechanisms cooperating intimately with adaptive immunity. Local generation of secretory IgA (SIgA) constitutes the largest humoral immune system of the body. Secretory antibodies function both by performing antigen exclusion at mucosal surfaces and by virus and endotoxin neutralization within epithelial cells without causing tissue damage. SIgA is thus persistently containing commensal bacteria outside the epithelial barrier but can also target invasion of pathogens and penetration of harmful antigens. Resistance to toxin-producing bacteria such as Vibrio cholerae and enterotoxigenic Escherichia coli appears to depend largely on SIgA, and so does herd protection against horizontal faecal-oral spread of enteric pathogens under naïve or immunized conditions--with a substantial innate impact both on cross-reactivity and memory. Like natural infections, live mucosal vaccines or adequate combinations of non-replicating vaccines and mucosal adjuvants, give rise not only to SIgA antibodies but also to longstanding serum IgG and IgA responses. However, there is considerably disparity with regard to migration of memory/effector cells from mucosal inductive sites to secretory effector sites and systemic immune organs. Also, although immunological memory is generated after mucosal priming, this may be masked by a self-limiting response protecting the inductive lymphoid tissue in the gut. The intranasal route of vaccine application targeting nasopharynx-associated lymphoid tissue may be more advantageous for certain infections, but only if successful stimulation is achieved without the use of toxic adjuvants that might reach the central nervous system. The degree of protection obtained after mucosal vaccination ranges from reduction of symptoms to complete inhibition of re-infection. In this scenario, it is often difficult to determine the relative importance of SIgA versus serum antibodies, but infection models in knockout mice strongly support the notion that SIgA exerts a decisive role in protection and cross-protection against a variety of infectious agents. Nevertheless, relatively few mucosal vaccines have been approved for human use, and more basic work is needed in vaccine and adjuvant design, including particulate or live-vectored combinations.
Collapse
Affiliation(s)
- Per Brandtzaeg
- Laboratory for Immunohistochemistry and Immunopathology, Institute and Department of Pathology, University of Oslo, Rikshospitalet-Radiumhospitalet Medical Centre, N-0027 Oslo, Norway.
| |
Collapse
|