1
|
Liaci AM, Chandra N, Vodnala SM, Strebl M, Kumar P, Pfenning V, Bachmann P, Caraballo R, Chai W, Johansson E, Elofsson M, Feizi T, Liu Y, Stehle T, Arnberg N. Extended receptor repertoire of an adenovirus associated with human obesity. PLoS Pathog 2025; 21:e1012892. [PMID: 39883726 PMCID: PMC11813153 DOI: 10.1371/journal.ppat.1012892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 02/11/2025] [Accepted: 01/07/2025] [Indexed: 02/01/2025] Open
Abstract
Human adenovirus type 36 (HAdV-D36) has been putatively linked to obesity in animals and has been associated with obesity in humans in some but not all studies. Despite extensive epidemiological research there is limited information about its receptor profile. We investigated the receptor portfolio of HAdV-D36 using a combined structural biology and virology approach. The HAdV-D36 fiber knob domain (FK), which mediates the primary attachment of many HAdVs to host cells, has a significantly elongated DG loop that alters known binding interfaces for established adenovirus receptors such as the coxsackie- and adenovirus receptor (CAR) and CD46. Our data suggest that HAdV-D36 attaches to host cells using a versatile receptor pool comprising sialic acid-containing glycans and CAR. Sialic acids are recognized at the same binding site used by other HAdVs of species D such as HAdV-D37. Using glycan microarrays, we demonstrate that HAdV-D36 displays a binding preference for glycans containing a rare sialic acid variant, 4-O,5-N-diacetylneuraminic acid, over the more common 5-N-acetylneuraminic acid. To date, this sialic acid variant has not been detected in humans, although it can be synthesized by various animal species, including a range of domestic and livestock animals. Taken together, our results indicate that HAdV-D36 has evolved to recognize a specialized set of primary attachment receptors that are different from known HAdV types and coincides with a unique host range and pathogenicity profile.
Collapse
Affiliation(s)
- A. Manuel Liaci
- Interfaculty Institute of Biochemistry, University of Tuebingen: Eberhard Karls Universitat Tubingen, Tuebingen, Germany
| | - Naresh Chandra
- Department of Clinical Microbiology, Umeå University, Umeå, Sweden
| | | | - Michael Strebl
- Interfaculty Institute of Biochemistry, University of Tuebingen: Eberhard Karls Universitat Tubingen, Tuebingen, Germany
| | - Pravin Kumar
- Department of Clinical Microbiology, Umeå University, Umeå, Sweden
| | - Vanessa Pfenning
- Interfaculty Institute of Biochemistry, University of Tuebingen: Eberhard Karls Universitat Tubingen, Tuebingen, Germany
| | - Paul Bachmann
- Interfaculty Institute of Biochemistry, University of Tuebingen: Eberhard Karls Universitat Tubingen, Tuebingen, Germany
| | - Rémi Caraballo
- Umeå Centre for Microbial Research, Umeå University, Umeå, Sweden
- Department of Chemistry, Umeå University, Umeå, Sweden
| | - Wengang Chai
- Glycosciences Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Emil Johansson
- Umeå Centre for Microbial Research, Umeå University, Umeå, Sweden
- Department of Chemistry, Umeå University, Umeå, Sweden
| | - Mikael Elofsson
- Umeå Centre for Microbial Research, Umeå University, Umeå, Sweden
- Department of Chemistry, Umeå University, Umeå, Sweden
| | - Ten Feizi
- Glycosciences Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Yan Liu
- Glycosciences Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Thilo Stehle
- Interfaculty Institute of Biochemistry, University of Tuebingen: Eberhard Karls Universitat Tubingen, Tuebingen, Germany
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Niklas Arnberg
- Department of Clinical Microbiology, Umeå University, Umeå, Sweden
- Umeå Centre for Microbial Research, Umeå University, Umeå, Sweden
| |
Collapse
|
2
|
Wang X, Hetzel M, Zhang W, Ehrhardt A, Bayer W. Comparative analysis of the impact of 40 adenovirus types on dendritic cell activation and CD8 + T cell proliferation capacity for the identification of favorable immunization vector candidates. Front Immunol 2023; 14:1286622. [PMID: 37915567 PMCID: PMC10616870 DOI: 10.3389/fimmu.2023.1286622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 10/04/2023] [Indexed: 11/03/2023] Open
Abstract
For the development of new adenovirus (AdV)-based vectors, it is important to understand differences in immunogenicity. In a side-by-side in vitro analysis, we evaluated the effect of 40 AdV types covering human AdV (HAdV) species A through G on the expression of 11 activation markers and the secretion of 12 cytokines by AdV-transduced dendritic cells, and the effect on CD8+ T cell proliferation capacity. We found that the expression of activation markers and cytokines differed widely between the different HAdV types, and many types were able to significantly impair the proliferation capacity of CD8+ T cells. Univariate and multivariate regression analyses suggested an important role of type I interferons in mediating this suppression of CD8+ T cells, which we confirmed experimentally in a proliferation assay using a type I interferon receptor blocking antibody. Using Bayesian statistics, we calculated a prediction model that suggests HAdV types HAdV-C1, -D8, -B7, -F41, -D33, -C2, -A31, -B3 and -D65 as the most favorable candidates for vaccine vector development.
Collapse
Affiliation(s)
- Xiaoyan Wang
- Institute for Virology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Mario Hetzel
- Institute for Virology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Wenli Zhang
- Virology and Microbiology, Center for Biomedical Education and Research (ZBAF), Faculty of Health, Witten/Herdecke University, Witten, Germany
| | - Anja Ehrhardt
- Virology and Microbiology, Center for Biomedical Education and Research (ZBAF), Faculty of Health, Witten/Herdecke University, Witten, Germany
| | - Wibke Bayer
- Institute for Virology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| |
Collapse
|
3
|
Persson BD, John L, Rafie K, Strebl M, Frängsmyr L, Ballmann MZ, Mindler K, Havenga M, Lemckert A, Stehle T, Carlson LA, Arnberg N. Human species D adenovirus hexon capsid protein mediates cell entry through a direct interaction with CD46. Proc Natl Acad Sci U S A 2021; 118:e2020732118. [PMID: 33384338 PMCID: PMC7826407 DOI: 10.1073/pnas.2020732118] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Human adenovirus species D (HAdV-D) types are currently being explored as vaccine vectors for coronavirus disease 2019 (COVID-19) and other severe infectious diseases. The efficacy of such vector-based vaccines depends on functional interactions with receptors on host cells. Adenoviruses of different species are assumed to enter host cells mainly by interactions between the knob domain of the protruding fiber capsid protein and cellular receptors. Using a cell-based receptor-screening assay, we identified CD46 as a receptor for HAdV-D56. The function of CD46 was validated in infection experiments using cells lacking and overexpressing CD46, and by competition infection experiments using soluble CD46. Remarkably, unlike HAdV-B types that engage CD46 through interactions with the knob domain of the fiber protein, HAdV-D types infect host cells through a direct interaction between CD46 and the hexon protein. Soluble hexon proteins (but not fiber knob) inhibited HAdV-D56 infection, and surface plasmon analyses demonstrated that CD46 binds to HAdV-D hexon (but not fiber knob) proteins. Cryoelectron microscopy analysis of the HAdV-D56 virion-CD46 complex confirmed the interaction and showed that CD46 binds to the central cavity of hexon trimers. Finally, soluble CD46 inhibited infection by 16 out of 17 investigated HAdV-D types, suggesting that CD46 is an important receptor for a large group of adenoviruses. In conclusion, this study identifies a noncanonical entry mechanism used by human adenoviruses, which adds to the knowledge of adenovirus biology and can also be useful for development of adenovirus-based vaccine vectors.
Collapse
Affiliation(s)
- B David Persson
- Department of Clinical Microbiology, Division of Virology, Umeå University, SE-90185 Umeå, Sweden
- Laboratory for Molecular Infection Medicine Sweden, Umeå University, SE-90185 Umeå, Sweden
| | - Lijo John
- Department of Clinical Microbiology, Division of Virology, Umeå University, SE-90185 Umeå, Sweden
- Laboratory for Molecular Infection Medicine Sweden, Umeå University, SE-90185 Umeå, Sweden
| | - Karim Rafie
- Laboratory for Molecular Infection Medicine Sweden, Umeå University, SE-90185 Umeå, Sweden
- Wallenberg Centre for Molecular Medicine, Umeå University, SE-90187 Umeå, Sweden
- Department of Medical Biochemistry, Umeå University, SE-90187 Umeå, Sweden
| | - Michael Strebl
- Interfaculty Institute of Biochemistry, The University of Tübingen, D-72076 Tübingen, Germany
| | - Lars Frängsmyr
- Department of Clinical Microbiology, Division of Virology, Umeå University, SE-90185 Umeå, Sweden
- Laboratory for Molecular Infection Medicine Sweden, Umeå University, SE-90185 Umeå, Sweden
| | | | - Katja Mindler
- Interfaculty Institute of Biochemistry, The University of Tübingen, D-72076 Tübingen, Germany
| | - Menzo Havenga
- Batavia Biosciences, 2333 CL Leiden, The Netherlands
| | | | - Thilo Stehle
- Interfaculty Institute of Biochemistry, The University of Tübingen, D-72076 Tübingen, Germany
| | - Lars-Anders Carlson
- Laboratory for Molecular Infection Medicine Sweden, Umeå University, SE-90185 Umeå, Sweden
- Wallenberg Centre for Molecular Medicine, Umeå University, SE-90187 Umeå, Sweden
- Department of Medical Biochemistry, Umeå University, SE-90187 Umeå, Sweden
| | - Niklas Arnberg
- Department of Clinical Microbiology, Division of Virology, Umeå University, SE-90185 Umeå, Sweden;
- Laboratory for Molecular Infection Medicine Sweden, Umeå University, SE-90185 Umeå, Sweden
| |
Collapse
|
4
|
Species D Adenoviruses as Oncolytic Viral Vectors. Viruses 2020; 12:v12121399. [PMID: 33291224 PMCID: PMC7762200 DOI: 10.3390/v12121399] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/01/2020] [Accepted: 12/03/2020] [Indexed: 12/18/2022] Open
Abstract
Oncolytic adenoviruses (Ad) have shown promising results in the therapeutic treatment of cancer. Ad type 5 (Ad5) is the most extensively utilized Ad type. However, several limitations exist to using Ad5 as an oncolytic virus, including high levels of anti-Ad5 neutralizing antibodies in the population, binding of the Ad5 hexon to blood coagulation factor X leading to liver sequestration and toxicity, and reduced expression of the primary receptor CAR on many tumors. Here, we use in vitro methods to explore the oncolytic potential of four alternative Ad types (Ad26, 28, 45, and 48) belonging to the species D Ad subgroup and developed replication-competent species D Ads expressing the human sodium iodide symporter protein (hNIS) for combination radiovirotherapy. We evaluated the species D Ad vectors transduction, replication, cytotoxicity, and gene expression in six different cancer cell lines. Species D Ads showed the greatest transduction and cytotoxic killing in the SKBR3 breast cancer cells, followed by 293, A549, and HepG2 cells, however the cytotoxicity was less than the wild type Ad5 virus. In contrast, species D Ads showed limited transduction and cytotoxicity in the Hela and SKOV3 cancer cell lines. These species D Ad vectors also successfully expressed the hNIS gene during infection leading to increased iodide uptake in multiple cancer cell lines. These results, the low seroprevalence of anti-species D antibodies, and the lack of binding to blood coagulation FX, support further exploration of species D Ads as alternative oncolytic adenoviruses against multiple types of cancer.
Collapse
|
5
|
Sasso E, D'Alise AM, Zambrano N, Scarselli E, Folgori A, Nicosia A. New viral vectors for infectious diseases and cancer. Semin Immunol 2020; 50:101430. [PMID: 33262065 DOI: 10.1016/j.smim.2020.101430] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 10/23/2020] [Accepted: 11/16/2020] [Indexed: 12/12/2022]
Abstract
Since the discovery in 1796 by Edward Jenner of vaccinia virus as a way to prevent and finally eradicate smallpox, the concept of using a virus to fight another virus has evolved into the current approaches of viral vectored genetic vaccines. In recent years, key improvements to the vaccinia virus leading to a safer version (Modified Vaccinia Ankara, MVA) and the discovery that some viruses can be used as carriers of heterologous genes encoding for pathological antigens of other infectious agents (the concept of 'viral vectors') has spurred a new wave of clinical research potentially providing for a solution for the long sought after vaccines against major diseases such as HIV, TB, RSV and Malaria, or emerging infectious diseases including those caused by filoviruses and coronaviruses. The unique ability of some of these viral vectors to stimulate the cellular arm of the immune response and, most importantly, T lymphocytes with cell killing activity, has also reawakened the interest toward developing therapeutic vaccines against chronic infectious diseases and cancer. To this end, existing vectors such as those based on Adenoviruses have been improved in immunogenicity and efficacy. Along the same line, new vectors that exploit viruses such as Vesicular Stomatitis Virus (VSV), Measles Virus (MV), Lymphocytic choriomeningitis virus (LCMV), cytomegalovirus (CMV), and Herpes Simplex Virus (HSV), have emerged. Furthermore, technological progress toward modifying their genome to render some of these vectors incompetent for replication has increased confidence toward their use in infant and elderly populations. Lastly, their production process being the same for every product has made viral vectored vaccines the technology of choice for rapid development of vaccines against emerging diseases and for 'personalised' cancer vaccines where there is an absolute need to reduce time to the patient from months to weeks or days. Here we review the recent developments in viral vector technologies, focusing on novel vectors based on primate derived Adenoviruses and Poxviruses, Rhabdoviruses, Paramixoviruses, Arenaviruses and Herpesviruses. We describe the rationale for, immunologic mechanisms involved in, and design of viral vectored gene vaccines under development and discuss the potential utility of these novel genetic vaccine approaches in eliciting protection against infectious diseases and cancer.
Collapse
Affiliation(s)
- Emanuele Sasso
- Nouscom srl, Via di Castel Romano 100, 00128 Rome, Italy; Ceinge-Biotecnologie Avanzate S.C. A.R.L., via Gaetano Salvatore 486, 80145 Naples, Italy.
| | | | - Nicola Zambrano
- Ceinge-Biotecnologie Avanzate S.C. A.R.L., via Gaetano Salvatore 486, 80145 Naples, Italy; Department of Molecular Medicine and Medical Biotechnology, University Federico II, Via Pansini 5, 80131 Naples, Italy.
| | | | | | - Alfredo Nicosia
- Ceinge-Biotecnologie Avanzate S.C. A.R.L., via Gaetano Salvatore 486, 80145 Naples, Italy; Department of Molecular Medicine and Medical Biotechnology, University Federico II, Via Pansini 5, 80131 Naples, Italy.
| |
Collapse
|
6
|
Sayedahmed EE, Elkashif A, Alhashimi M, Sambhara S, Mittal SK. Adenoviral Vector-Based Vaccine Platforms for Developing the Next Generation of Influenza Vaccines. Vaccines (Basel) 2020; 8:vaccines8040574. [PMID: 33019589 PMCID: PMC7712206 DOI: 10.3390/vaccines8040574] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 09/06/2020] [Accepted: 09/17/2020] [Indexed: 12/20/2022] Open
Abstract
Ever since the discovery of vaccines, many deadly diseases have been contained worldwide, ultimately culminating in the eradication of smallpox and polio, which represented significant medical achievements in human health. However, this does not account for the threat influenza poses on public health. The currently licensed seasonal influenza vaccines primarily confer excellent strain-specific protection. In addition to the seasonal influenza viruses, the emergence and spread of avian influenza pandemic viruses such as H5N1, H7N9, H7N7, and H9N2 to humans have highlighted the urgent need to adopt a new global preparedness for an influenza pandemic. It is vital to explore new strategies for the development of effective vaccines for pandemic and seasonal influenza viruses. The new vaccine approaches should provide durable and broad protection with the capability of large-scale vaccine production within a short time. The adenoviral (Ad) vector-based vaccine platform offers a robust egg-independent production system for manufacturing large numbers of influenza vaccines inexpensively in a short timeframe. In this review, we discuss the progress in the development of Ad vector-based influenza vaccines and their potential in designing a universal influenza vaccine.
Collapse
Affiliation(s)
- Ekramy E. Sayedahmed
- Department of Comparative Pathobiology, Purdue Institute for Immunology, Inflammation and Infectious Disease, Purdue University Center for Cancer Research, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA; (E.E.S.); (A.E.); (M.A.)
| | - Ahmed Elkashif
- Department of Comparative Pathobiology, Purdue Institute for Immunology, Inflammation and Infectious Disease, Purdue University Center for Cancer Research, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA; (E.E.S.); (A.E.); (M.A.)
| | - Marwa Alhashimi
- Department of Comparative Pathobiology, Purdue Institute for Immunology, Inflammation and Infectious Disease, Purdue University Center for Cancer Research, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA; (E.E.S.); (A.E.); (M.A.)
| | - Suryaprakash Sambhara
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA
- Correspondence: (S.S.); (S.K.M.)
| | - Suresh K. Mittal
- Department of Comparative Pathobiology, Purdue Institute for Immunology, Inflammation and Infectious Disease, Purdue University Center for Cancer Research, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA; (E.E.S.); (A.E.); (M.A.)
- Correspondence: (S.S.); (S.K.M.)
| |
Collapse
|
7
|
Napolitano F, Merone R, Abbate A, Ammendola V, Horncastle E, Lanzaro F, Esposito M, Contino AM, Sbrocchi R, Sommella A, Duncan JD, Hinds J, Urbanowicz RA, Lahm A, Colloca S, Folgori A, Ball JK, Nicosia A, Wizel B, Capone S, Vitelli A. A next generation vaccine against human rabies based on a single dose of a chimpanzee adenovirus vector serotype C. PLoS Negl Trop Dis 2020; 14:e0008459. [PMID: 32667913 PMCID: PMC7363076 DOI: 10.1371/journal.pntd.0008459] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 06/05/2020] [Indexed: 12/25/2022] Open
Abstract
Rabies, caused by RNA viruses in the Genus Lyssavirus, is the most fatal of all infectious diseases. This neglected zoonosis remains a major public health problem in developing countries, causing the death of an estimated 25,000-159,000 people each year, with more than half of them in children. The high incidence of human rabies in spite of effective vaccines is mainly linked to the lack of compliance with the complicated administration schedule, inadequacies of the community public health system for local administration by the parenteral route and the overall costs of the vaccine. The goal of our work was the development of a simple, affordable and effective vaccine strategy to prevent human rabies virus infection. This next generation vaccine is based on a replication-defective chimpanzee adenovirus vector belonging to group C, ChAd155-RG, which encodes the rabies glycoprotein (G). We demonstrate here that a single dose of this vaccine induces protective efficacy in a murine model of rabies challenge and elicits strong and durable neutralizing antibody responses in vaccinated non-human primates. Importantly, we demonstrate that one dose of a commercial rabies vaccine effectively boosts the neutralizing antibody responses induced by ChAd155-RG in vaccinated monkeys, showing the compatibility of the novel vectored vaccine with the current post-exposure prophylaxis in the event of rabies virus exposure. Finally, we demonstrate that antibodies induced by ChAd155-RG can also neutralize European bat lyssaviruses 1 and 2 (EBLV-1 and EBLV-2) found in bat reservoirs.
Collapse
Affiliation(s)
| | | | | | | | - Emma Horncastle
- Wolfson Centre for Global Virus Infections, University of Nottingham, Nottingham, United Kingdom
- School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | | | | | | | | | | | - Joshua D. Duncan
- Wolfson Centre for Global Virus Infections, University of Nottingham, Nottingham, United Kingdom
- School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Jospeh Hinds
- Wolfson Centre for Global Virus Infections, University of Nottingham, Nottingham, United Kingdom
- School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Richard A. Urbanowicz
- Wolfson Centre for Global Virus Infections, University of Nottingham, Nottingham, United Kingdom
- School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | | | | | | | - Jonathan K. Ball
- Wolfson Centre for Global Virus Infections, University of Nottingham, Nottingham, United Kingdom
- School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Alfredo Nicosia
- Department of Molecular Medicine and Medical Biotechnology, University Federico II, Naples, Italy
| | - Benjamin Wizel
- GSK Vaccines, Rockville, Maryland, United States of America
| | | | | |
Collapse
|
8
|
Coughlan L. Factors Which Contribute to the Immunogenicity of Non-replicating Adenoviral Vectored Vaccines. Front Immunol 2020; 11:909. [PMID: 32508823 PMCID: PMC7248264 DOI: 10.3389/fimmu.2020.00909] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 04/20/2020] [Indexed: 01/12/2023] Open
Abstract
Adenoviral vectors are a safe and potently immunogenic vaccine delivery platform. Non-replicating Ad vectors possess several attributes which make them attractive vaccines for infectious disease, including their capacity for high titer growth, ease of manipulation, safety, and immunogenicity in clinical studies, as well as their compatibility with clinical manufacturing and thermo-stabilization procedures. In general, Ad vectors are immunogenic vaccines, which elicit robust transgene antigen-specific cellular (namely CD8+ T cells) and/or humoral immune responses. A large number of adenoviruses isolated from humans and non-human primates, which have low seroprevalence in humans, have been vectorized and tested as vaccines in animal models and humans. However, a distinct hierarchy of immunological potency has been identified between diverse Ad vectors, which unfortunately limits the potential use of many vectors which have otherwise desirable manufacturing characteristics. The precise mechanistic factors which underlie the profound disparities in immunogenicity are not clearly defined and are the subject of ongoing, detailed investigation. It has been suggested that a combination of factors contribute to the potent immunogenicity of particular Ad vectors, including the magnitude and duration of vaccine antigen expression following immunization. Furthermore, the excessive induction of Type I interferons by some Ad vectors has been suggested to impair transgene expression levels, dampening subsequent immune responses. Therefore, the induction of balanced, but not excessive stimulation of innate signaling is optimal. Entry factor binding or receptor usage of distinct Ad vectors can also affect their in vivo tropism following administration by different routes. The abundance and accessibility of innate immune cells and/or antigen-presenting cells at the site of injection contributes to early innate immune responses to Ad vaccination, affecting the outcome of the adaptive immune response. Although a significant amount of information exists regarding the tropism determinants of the common human adenovirus type-5 vector, very little is known about the receptor usage and tropism of rare species or non-human Ad vectors. Increased understanding of how different facets of the host response to Ad vectors contribute to their immunological potency will be essential for the development of optimized and customized Ad vaccine platforms for specific diseases.
Collapse
|
9
|
Gao J, Mese K, Bunz O, Ehrhardt A. State‐of‐the‐art human adenovirus vectorology for therapeutic approaches. FEBS Lett 2019; 593:3609-3622. [DOI: 10.1002/1873-3468.13691] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/16/2019] [Accepted: 11/18/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Jian Gao
- Faculty of Health Centre for Biomedical Education and Research (ZBAF) School of Human Medicine Institute of Virology and Microbiology Witten/Herdecke University Germany
| | - Kemal Mese
- Faculty of Health Centre for Biomedical Education and Research (ZBAF) School of Human Medicine Institute of Virology and Microbiology Witten/Herdecke University Germany
| | - Oskar Bunz
- Faculty of Health Centre for Biomedical Education and Research (ZBAF) School of Human Medicine Institute of Virology and Microbiology Witten/Herdecke University Germany
| | - Anja Ehrhardt
- Faculty of Health Centre for Biomedical Education and Research (ZBAF) School of Human Medicine Institute of Virology and Microbiology Witten/Herdecke University Germany
| |
Collapse
|
10
|
Darrah PA, DiFazio RM, Maiello P, Gideon HP, Myers AJ, Rodgers MA, Hackney JA, Lindenstrom T, Evans T, Scanga CA, Prikhodko V, Andersen P, Lin PL, Laddy D, Roederer M, Seder RA, Flynn JL. Boosting BCG with proteins or rAd5 does not enhance protection against tuberculosis in rhesus macaques. NPJ Vaccines 2019; 4:21. [PMID: 31149352 PMCID: PMC6538611 DOI: 10.1038/s41541-019-0113-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 05/03/2019] [Indexed: 12/31/2022] Open
Abstract
Tuberculosis (TB) is the leading cause of death from infection worldwide. The only approved vaccine, BCG, has variable protective efficacy against pulmonary TB, the transmissible form of the disease. Therefore, improving this efficacy is an urgent priority. This study assessed whether heterologous prime-boost vaccine regimens in which BCG priming is boosted with either (i) protein and adjuvant (M72 plus AS01E or H56 plus CAF01) delivered intramuscularly (IM), or (ii) replication-defective recombinant adenovirus serotype 5 (Ad5) expressing various Mycobacterium tuberculosis (Mtb) antigens (Ad5(TB): M72, ESAT-6/Ag85b, or ESAT-6/Rv1733/Rv2626/RpfD) administered simultaneously by IM and aerosol (AE) routes, could enhance blood- and lung-localized T-cell immunity and improve protection in a nonhuman primate (NHP) model of TB infection. Ad5(TB) vaccines administered by AE/IM routes following BCG priming elicited ~10-30% antigen-specific CD4 and CD8 T-cell multifunctional cytokine responses in bronchoalveolar lavage (BAL) but did not provide additional protection compared to BCG alone. Moreover, AE administration of an Ad5(empty) control vector after BCG priming appeared to diminish protection induced by BCG. Boosting BCG by IM immunization of M72/AS01E or H56:CAF01 elicited ~0.1-0.3% antigen-specific CD4 cytokine responses in blood with only a transient increase of ~0.5-1% in BAL; these vaccine regimens also failed to enhance BCG-induced protection. Taken together, this study shows that boosting BCG with protein/adjuvant or Ad-based vaccines using these antigens, by IM or IM/AE routes, respectively, do not enhance protection against primary infection compared with BCG alone, in the highly susceptible rhesus macaque model of tuberculosis.
Collapse
Affiliation(s)
- Patricia A Darrah
- 1Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD USA
| | - Robert M DiFazio
- 2Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - Pauline Maiello
- 2Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - Hannah P Gideon
- 2Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - Amy J Myers
- 2Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - Mark A Rodgers
- 2Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - Joshua A Hackney
- 1Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD USA
| | - Thomas Lindenstrom
- 3Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | | | - Charles A Scanga
- 2Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | | | - Peter Andersen
- 3Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Philana Ling Lin
- 5Department of Pediatrics, Children's Hospital of the University of Pittsburgh of UPMC, Pittsburgh, PA USA
| | | | - Mario Roederer
- 1Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD USA
| | - Robert A Seder
- 1Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD USA
| | - JoAnne L Flynn
- 2Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| |
Collapse
|
11
|
Mennechet FJD, Paris O, Ouoba AR, Salazar Arenas S, Sirima SB, Takoudjou Dzomo GR, Diarra A, Traore IT, Kania D, Eichholz K, Weaver EA, Tuaillon E, Kremer EJ. A review of 65 years of human adenovirus seroprevalence. Expert Rev Vaccines 2019; 18:597-613. [PMID: 31132024 DOI: 10.1080/14760584.2019.1588113] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Introduction: Human adenovirus (HAdV)-derived vectors have been used in numerous pre-clinical and clinical trials during the last 40 years. Current research in HAdV-based vaccines focuses on improving transgene immunogenicity and safety. Because pre-existing humoral immunity against HAdV types correlate with reduced vaccine efficacy and safety, many groups are exploring the development of HAdV types vectors with lower seroprevalence. However, global seroepidemiological data are incomplete. Areas covered: The goal of this review is to centralize 65 years of research on (primarily) HAdV epidemiology. After briefly addressing adenovirus biology, we chronical HAdV seroprevalence studies and highlight major milestones. Finally, we analyze data from about 50 studies with respect to HAdVs types that are currently used in the clinic, or are in the developmental pipeline. Expert opinion: Vaccination is among the most efficient tools to prevent infectious disease. HAdV-based vaccines have undeniable potential, but optimization is needed and antivector immunity remains a challenge if the same vectors are to be administrated to different populations. Here, we identify gaps in our knowledge and the need for updated worldwide epidemiological data.
Collapse
Affiliation(s)
- Franck J D Mennechet
- a Institut de Génétique Moléculaire de Montpellier , University of Montpellier - CNRS , Montpellier , France
| | - Océane Paris
- a Institut de Génétique Moléculaire de Montpellier , University of Montpellier - CNRS , Montpellier , France
| | - Aline Raissa Ouoba
- a Institut de Génétique Moléculaire de Montpellier , University of Montpellier - CNRS , Montpellier , France.,b UMR 1058, Pathogenesis and Control of Chronic Infections , INSERM - University of Montpellier - Establishment Français du Sang - Centre Hospitalier Universitaire de Montpellier , Montpellier , France.,c Département des sciences et de la recherche clinique , Centre Muraz , Bobo-Dioulasso , Burkina Faso
| | - Sofia Salazar Arenas
- a Institut de Génétique Moléculaire de Montpellier , University of Montpellier - CNRS , Montpellier , France
| | - Sodiomon B Sirima
- d Centre National de Recherche et de Formation sur le Paludisme , Ouagadougou , Burkina Faso.,e Groupe de Recherche Action en Santé (GRAS) , Ouagadougou , Burkina Faso
| | - Guy R Takoudjou Dzomo
- f Complexe Hospitalo Universitaire « Le Bon Samaritain » , N'Djamena , Republic of Chad
| | - Amidou Diarra
- d Centre National de Recherche et de Formation sur le Paludisme , Ouagadougou , Burkina Faso
| | - Isidore T Traore
- c Département des sciences et de la recherche clinique , Centre Muraz , Bobo-Dioulasso , Burkina Faso
| | - Dramane Kania
- c Département des sciences et de la recherche clinique , Centre Muraz , Bobo-Dioulasso , Burkina Faso
| | - Karsten Eichholz
- a Institut de Génétique Moléculaire de Montpellier , University of Montpellier - CNRS , Montpellier , France
| | - Eric A Weaver
- g University of Nebraska-Lincoln, School of Biological Sciences , Lincoln , NE , USA
| | - Edouard Tuaillon
- b UMR 1058, Pathogenesis and Control of Chronic Infections , INSERM - University of Montpellier - Establishment Français du Sang - Centre Hospitalier Universitaire de Montpellier , Montpellier , France
| | - Eric J Kremer
- a Institut de Génétique Moléculaire de Montpellier , University of Montpellier - CNRS , Montpellier , France
| |
Collapse
|
12
|
Goyvaerts C, Breckpot K. The Journey of in vivo Virus Engineered Dendritic Cells From Bench to Bedside: A Bumpy Road. Front Immunol 2018; 9:2052. [PMID: 30254636 PMCID: PMC6141723 DOI: 10.3389/fimmu.2018.02052] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 08/20/2018] [Indexed: 12/13/2022] Open
Abstract
Dendritic cells (DCs) are recognized as highly potent antigen-presenting cells that are able to stimulate cytotoxic T lymphocyte (CTL) responses with antitumor activity. Consequently, DCs have been explored as cellular vaccines in cancer immunotherapy. To that end, DCs are modified with tumor antigens to enable presentation of antigen-derived peptides to CTLs. In this review we discuss the use of viral vectors for in situ modification of DCs, focusing on their clinical applications as anticancer vaccines. Among the viral vectors discussed are those derived from viruses belonging to the families of the Poxviridae, Adenoviridae, Retroviridae, Togaviridae, Paramyxoviridae, and Rhabdoviridae. We will further shed light on how the combination of viral vector-based vaccination with T-cell supporting strategies will bring this strategy to the next level.
Collapse
|
13
|
Improved Induction of Anti-Melanoma T Cells by Adenovirus-5/3 Fiber Modification to Target Human DCs. Vaccines (Basel) 2018; 6:vaccines6030042. [PMID: 30022005 PMCID: PMC6161112 DOI: 10.3390/vaccines6030042] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 07/12/2018] [Accepted: 07/16/2018] [Indexed: 12/14/2022] Open
Abstract
To mount a strong anti-tumor immune response, non T cell inflamed (cold) tumors may require combination treatment encompassing vaccine strategies preceding checkpoint inhibition. In vivo targeted delivery of tumor-associated antigens (TAA) to dendritic cells (DCs), relying on the natural functions of primary DCs in situ, represents an attractive vaccination strategy. In this study we made use of a full-length MART-1 expressing C/B-chimeric adenoviral vector, consisting of the Ad5 capsid and the Ad3 knob (Ad5/3), which we previously showed to selectively transduce DCs in human skin and lymph nodes. Our data demonstrate that chimeric Ad5/3 vectors encoding TAA, and able to target human DCs in situ, can be used to efficiently induce expansion of functional tumor-specific CD8+ effector T cells, either from a naïve T cell pool or from previously primed T cells residing in the melanoma-draining sentinel lymph nodes (SLN). These data support the use of Ad3-knob containing viruses as vaccine vehicles for in vivo delivery. “Off-the-shelf” DC-targeted Ad vaccines encoding TAA could clearly benefit future immunotherapeutic approaches.
Collapse
|
14
|
Native and engineered tropism of vectors derived from a rare species D adenovirus serotype 43. Oncotarget 2018; 7:53414-53429. [PMID: 27462785 PMCID: PMC5288196 DOI: 10.18632/oncotarget.10800] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 07/13/2016] [Indexed: 02/03/2023] Open
Abstract
Unique molecular properties of species D adenoviruses (Ads)—the most diverse yet underexplored group of Ads—have been used to develop improved gene vectors. The low seroprevalence in humans of adenovirus serotype 43 (Ad43), an otherwise unstudied species D Ad, identified this rare serotype as an attractive new human gene therapy vector platform. Thus, in this study we wished to assess biological properties of Ad43 essential to its vectorization. We found that (1) Ad43 virions do not bind blood coagulation factor X and cause low random transduction upon vascular delivery; (2) they clear host tissues more quickly than do traditionally used Ad5 vectors; (3) Ad43 uses CD46 as primary receptor; (4) Ad43 can use integrins as alternative primary receptors. As the first step toward vectorization of Ad43, we demonstrated that the primary receptor specificity of the Ad43 fiber can be altered to achieve infection via Her2, an established oncotarget. Whereas this modification required use of the Ad5 fiber shaft, the presence of this domain in chimeric virions did not make them susceptible for neutralization by anti-Ad5 antibodies.
Collapse
|
15
|
Nidetz NF, Gallagher TM, Wiethoff CM. Inhibition of type I interferon responses by adenovirus serotype-dependent Gas6 binding. Virology 2017; 515:150-157. [PMID: 29288958 DOI: 10.1016/j.virol.2017.12.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 12/14/2017] [Accepted: 12/15/2017] [Indexed: 10/18/2022]
Abstract
The clinical use of many adenovirus vaccine vectors (AdVs) is limited by the presence of pre-existing antibodies in human populations, which prevent common AdVs from transducing cells and expressing immunogenic gene products. Rare serotype AdVs, such as HAdV-28D can bypass pre-existing immunity. However, rare AdVs stimulate high-levels of type I interferon (IFN), which suppresses antigenic gene expression and therefore limits immunogenicity. Recent studies identified Gas6 as a factor that connects enveloped viruses to host-cell receptor tyrosine kinases, in turn generating signaling cascades that antagonize type I IFN responses. We discovered that Gas6 bound to the fiber proteins of common AdV serotypes, such as HAdV-5C, with a higher affinity than rare HAd-28D fibers. AdV-associated Gas6 suppressed IFN production by common AdVs and enhanced long-term expression of AdV encoded genes. We hypothesize that rare AdV serotypes might be engineered to include Gas6 binding motifs, thereby generating novel vectors that are more effective.
Collapse
Affiliation(s)
- Natalie F Nidetz
- Loyola University Chicago, 2016 S. First Avenue, Maywood, IL 60153, USA.
| | - Tom M Gallagher
- Loyola University Chicago, 2016 S. First Avenue, Maywood, IL 60153, USA.
| | | |
Collapse
|
16
|
Duffy MR, Alonso-Padilla J, John L, Chandra N, Khan S, Ballmann MZ, Lipiec A, Heemskerk E, Custers J, Arnberg N, Havenga M, Baker AH, Lemckert A. Generation and characterization of a novel candidate gene therapy and vaccination vector based on human species D adenovirus type 56. J Gen Virol 2017; 99:135-147. [PMID: 29154744 DOI: 10.1099/jgv.0.000978] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The vectorization of rare human adenovirus (HAdV) types will widen our knowledge of this family and their interaction with cells, tissues and organs. In this study we focus on HAdV-56, a member of human Ad species D, and create ease-of-use cloning systems to generate recombinant HAdV-56 vectors carrying foreign genes. We present in vitro transduction profiles for HAdV-56 in direct comparison to the most commonly used HAdV-5-based vector. In vivo characterizations demonstrate that when it is delivered intravenously (i.v.) HAdV-56 mainly targets the spleen and, to a lesser extent, the lungs, whilst largely bypassing liver transduction in mice. HAdV-56 triggered robust inflammatory and cellular immune responses, with higher induction of IFNγ, TNFα, IL5, IL6, IP10, MCP1 and MIG1 compared to HAdV-5 following i.v. administration. We also investigated its potential as a vaccine vector candidate by performing prime immunizations in mice with HAdV-56 encoding luciferase (HAdV-56-Luc). Direct comparisons were made to HAdV-26, a highly potent human vaccine vector currently in phase II clinical trials. HAdV-56-Luc induced luciferase 'antigen'-specific IFNγ-producing cells and anti-HAdV-56 neutralizing antibodies in Balb/c mice, demonstrating a near identical profile to that of HAdV-26. Taken together, the data presented provides further insight into human Ad receptor/co-receptor usage, and the first report on HAdV-56 vectors and their potential for gene therapy and vaccine applications.
Collapse
Affiliation(s)
- Margaret R Duffy
- Batavia Biosciences BV, Leiden, The Netherlands.,Present address: Department of Oncology, University of Oxford, Oxford, UK
| | - Julio Alonso-Padilla
- Institute of Cardiovascular and Medical Sciences, College of Medicine, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK.,Present address: Barcelona Institute for Global Health (ISGlobal), Centre for Research in International Health (CRESIB), Hospital Clinic de Barcelona -University of Barcelona, Barcelona, Spain
| | - Lijo John
- Division of Virology, Department of Clinical Microbiology, Umeå University, Sweden
| | - Naresh Chandra
- Division of Virology, Department of Clinical Microbiology, Umeå University, Sweden
| | - Selina Khan
- Viral Vaccine Discovery and Early Development, Janssen Vaccines and Prevention BV, Leiden, The Netherlands
| | | | | | | | - Jerome Custers
- Viral Vaccine Discovery and Early Development, Janssen Vaccines and Prevention BV, Leiden, The Netherlands
| | - Niklas Arnberg
- Division of Virology, Department of Clinical Microbiology, Umeå University, Sweden
| | | | - Andrew H Baker
- Present address: Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK.,Institute of Cardiovascular and Medical Sciences, College of Medicine, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | | |
Collapse
|
17
|
Vitelli A, Folgori A, Scarselli E, Colloca S, Capone S, Nicosia A. Chimpanzee adenoviral vectors as vaccines - challenges to move the technology into the fast lane. Expert Rev Vaccines 2017; 16:1241-1252. [PMID: 29047309 DOI: 10.1080/14760584.2017.1394842] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION In recent years, replication-defective chimpanzee-derived adenoviruses have been extensively evaluated as genetic vaccines. These vectors share desirable properties with human adenoviruses like the broad tissue tropism and the ease of large-scale manufacturing. Additionally, chimpanzee adenoviruses have the advantage to overcome the negative impact of pre-existing anti-human adenovirus immunity. Areas covered: Here the authors review current pre-clinical research and clinical trials that utilize chimpanzee-derived adenoviral vectors as vaccines. A wealth of studies are ongoing to evaluate different vector backbones and administration routes with the aim of improving immune responses. The challenges associated with the identification of an optimal chimpanzee vector and immunization strategies for different immunological outcomes will be discussed. Expert commentary: The demonstration that chimpanzee adenoviruses can be safely used in humans has paved the way to the use of a whole new array of vectors of different serotypes. However, so far no predictive signature of vector immunity in humans has been identified. The high magnitude of T cell responses elicited by chimpanzee adenoviruses has allowed dissecting the qualitative aspects that may be important for protective immunity. Ultimately, only the results from the most clinically advanced products will help establish the efficacy of the vaccine vector platform in the field of disease prevention.
Collapse
Affiliation(s)
| | | | | | | | | | - Alfredo Nicosia
- a ReiThera , Rome , Italy.,c CEINGE , Naples , Italy.,d Department of Molecular Medicine and Medical Biotechnology , University of Naples Federico II , Naples , Italy
| |
Collapse
|
18
|
Zhang W, Ehrhardt A. Getting genetic access to natural adenovirus genomes to explore vector diversity. Virus Genes 2017; 53:675-683. [PMID: 28711987 DOI: 10.1007/s11262-017-1487-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 07/06/2017] [Indexed: 01/20/2023]
Abstract
Recombinant vectors based on the human adenovirus type 5 (HAdV5) have been developed and extensively used in preclinical and clinical studies for over 30 years. However, certain restrictions of HAdV5-based vectors have limited their clinical applications because they are rather inefficient in specifically transducing cells of therapeutic interest that lack the coxsackievirus and adenovirus receptor (CAR). Moreover, enhanced vector-associated toxicity and widespread preexisting immunity have been shown to significantly hamper the effectiveness of HAdV-5-mediated gene transfer. However, evolution of adenoviruses in the natural host is driving the generation of novel types with altered virulence, enhanced transmission, and altered tissue tropism. As a consequence, an increasing number of alternative adenovirus types were identified, which may represent a valuable resource for the development of novel vector types. Thus, researchers are focusing on the other naturally occurring adenovirus types, which are structurally similar but functionally different from HAdV5. To this end, several strategies have been devised for getting genetic access to adenovirus genomes, resulting in a new panel of adenoviral vectors. Importantly, these vectors were shown to have a host range different from HAdV5 and to escape the anti-HAdV5 immune response, thus underlining the great potential of this approach. In summary, this review provides a state-of-the-art overview of one essential step in adenoviral vector development.
Collapse
Affiliation(s)
- Wenli Zhang
- Department of Human Medicine, Faculty of Health, Institute of Virology and Microbiology, Center for Biomedical Education and Research (ZBAF), Witten/Herdecke University, 58453, Witten, Germany
| | - Anja Ehrhardt
- Department of Human Medicine, Faculty of Health, Institute of Virology and Microbiology, Center for Biomedical Education and Research (ZBAF), Witten/Herdecke University, 58453, Witten, Germany.
| |
Collapse
|
19
|
Xiang K, Ying G, Yan Z, Shanshan Y, Lei Z, Hongjun L, Maosheng S. Progress on adenovirus-vectored universal influenza vaccines. Hum Vaccin Immunother 2016; 11:1209-22. [PMID: 25876176 DOI: 10.1080/21645515.2015.1016674] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Influenza virus (IFV) infection causes serious health problems and heavy financial burdens each year worldwide. The classical inactivated influenza virus vaccine (IIVV) and live attenuated influenza vaccine (LAIV) must be updated regularly to match the new strains that evolve due to antigenic drift and antigenic shift. However, with the discovery of broadly neutralizing antibodies that recognize conserved antigens, and the CD8(+) T cell responses targeting viral internal proteins nucleoprotein (NP), matrix protein 1 (M1) and polymerase basic 1 (PB1), it is possible to develop a universal influenza vaccine based on the conserved hemagglutinin (HA) stem, NP, and matrix proteins. Recombinant adenovirus (rAd) is an ideal influenza vaccine vector because it has an ideal stability and safety profile, induces balanced humoral and cell-mediated immune responses due to activation of innate immunity, provides 'self-adjuvanting' activity, can mimic natural IFV infection, and confers seamless protection against mucosal pathogens. Moreover, this vector can be developed as a low-cost, rapid-response vaccine that can be quickly manufactured. Therefore, an adenovirus vector encoding conserved influenza antigens holds promise in the development of a universal influenza vaccine. This review will summarize the progress in adenovirus-vectored universal flu vaccines and discuss future novel approaches.
Collapse
Key Words
- ADCC, antibody-dependent cell-mediated cytotoxicity
- APC, antigen-presenting cell
- Ad: adenovirus
- CAR, Coxsackie-Adenovirus Receptor
- CTLs, cytotoxic T lymphocytes
- DC, lung dendritic cells
- DVD, drug–vaccine duo
- FcγRs, Fc receptors for IgG
- HA, hemagglutinin
- HDAd, helper-dependent adenoviral
- HEK293, human embryonic kidney 293 cell
- HI, hemagglutination inhibition
- HLA, human leukocyte antigen
- IF-γ, interferon-γ
- IFV, Influenza virus
- IIVV, inactivated influenza virus vaccine
- IL-2, interleukin-2
- ITRs, inverted terminal repeats
- LAIV, live attenuated influenza vaccine
- M1, matrix protein 1
- M2, matrix protein 2
- MHC-I, major histocompatibility complex class I
- NA, neuraminidase
- NP, nucleoprotein
- RCA, replication competent adenovirus
- VAERD, vaccine-associated enhanced respiratory disease
- adenovirus vector
- broadly neutralizing antibodies
- cellular immunity
- flu, influenza
- hemagglutinin
- humoral immunity
- influenza
- mAbs, monoclonal antibodies
- mucosal immunity
- rAd, recombinant adenovirus
- universal vaccine
Collapse
Affiliation(s)
- Kui Xiang
- a Department of Molecular Biology; Institute of Medical Biology; Chinese Academy of Medical Sciences; Peking Union Medical College ; Kunming , Yunnan , PR China
| | | | | | | | | | | | | |
Collapse
|
20
|
Abstract
Many nonhuman adenoviruses (AdVs) of simian, bovine, porcine, canine, ovine, murine, and fowl origin are being developed as gene delivery systems for recombinant vaccines and gene therapy applications. In addition to circumventing preexisting human AdV (HAdV) immunity, nonhuman AdV vectors utilize coxsackievirus-adenovirus receptor or other receptors for vector internalization, thereby expanding the range of cell types that can be targeted. Nonhuman AdV vectors also provide excellent platforms for veterinary vaccines. A specific nonhuman AdV vector when used in its species of origin could provide an excellent animal model for evaluating the vector efficacy and pathogenesis. These vectors are useful in prime–boost approaches with other AdV vectors or with other gene delivery systems including DNA immunization and viral or bacterial vectors. When multiple vector inoculations are required, nonhuman AdV vectors could supplement HAdV or other viral vectors.
Collapse
|
21
|
Development of Novel Adenoviral Vectors to Overcome Challenges Observed With HAdV-5-based Constructs. Mol Ther 2015; 24:6-16. [PMID: 26478249 PMCID: PMC4754553 DOI: 10.1038/mt.2015.194] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 10/07/2015] [Indexed: 12/23/2022] Open
Abstract
Recombinant vectors based on human adenovirus serotype 5 (HAdV-5) have been extensively studied in preclinical models and clinical trials over the past two decades. However, the thorough understanding of the HAdV-5 interaction with human subjects has uncovered major concerns about its product applicability. High vector-associated toxicity and widespread preexisting immunity have been shown to significantly impede the effectiveness of HAdV-5–mediated gene transfer. It is therefore that the in-depth knowledge attained working on HAdV-5 is currently being used to develop alternative vectors. Here, we provide a comprehensive overview of data obtained in recent years disqualifying the HAdV-5 vector for systemic gene delivery as well as novel strategies being pursued to overcome the limitations observed with particular emphasis on the ongoing vectorization efforts to obtain vectors based on alternative serotypes.
Collapse
|
22
|
A Dual-Modality Herpes Simplex Virus 2 Vaccine for Preventing Genital Herpes by Using Glycoprotein C and D Subunit Antigens To Induce Potent Antibody Responses and Adenovirus Vectors Containing Capsid and Tegument Proteins as T Cell Immunogens. J Virol 2015; 89:8497-509. [PMID: 26041292 DOI: 10.1128/jvi.01089-15] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 05/27/2015] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED We evaluated a genital herpes prophylactic vaccine containing herpes simplex virus 2 (HSV-2) glycoproteins C (gC2) and D (gD2) to stimulate humoral immunity and UL19 (capsid protein VP5) and UL47 (tegument protein VP13/14) as T cell immunogens. The HSV-2 gC2 and gD2 proteins were expressed in baculovirus, while the UL19 and UL47 genes were expressed from replication-defective adenovirus vectors. Adenovirus vectors containing UL19 and UL47 stimulated human and murine CD4(+) and CD8(+) T cell responses. Guinea pigs were either (i) mock immunized; (ii) immunized with gC2/gD2, with CpG and alum as adjuvants; (iii) immunized with the UL19/UL47 adenovirus vectors; or (iv) immunized with the combination of gC2/gD2-CpG/alum and the UL19/UL47 adenovirus vectors. Immunization with gC2/gD2 produced potent neutralizing antibodies, while UL19 and UL47 also stimulated antibody responses. After intravaginal HSV-2 challenge, the mock and UL19/UL47 adenovirus groups developed severe acute disease, while 2/8 animals in the gC2/gD2-only group and none in the combined group developed acute disease. No animals in the gC2/gD2 or combined group developed recurrent disease; however, 5/8 animals in each group had subclinical shedding of HSV-2 DNA, on 15/168 days for the gC2/gD2 group and 13/168 days for the combined group. Lumbosacral dorsal root ganglia were positive for HSV-2 DNA and latency-associated transcripts for 5/8 animals in the gC2/gD2 group and 2/8 animals in the combined group. None of the differences comparing the gC2/gD2-only group and the combined group were statistically significant. Therefore, adding the T cell immunogens UL19 and UL47 to the gC2/gD2 vaccine did not significantly reduce genital disease and vaginal HSV-2 DNA shedding compared with the excellent protection provided by gC2/gD2 in the guinea pig model. IMPORTANCE HSV-2 infection is a common cause of genital ulcer disease and a significant public health concern. Genital herpes increases the risk of transmission and acquisition of HIV-1 infection 3- to 4-fold. A herpes vaccine that prevents genital lesions and asymptomatic genital shedding will have a substantial impact on two epidemics, i.e., both the HSV-2 and HIV-1 epidemics. We previously reported that a vaccine containing HSV-2 glycoprotein C (gC2) and glycoprotein D (gD2) reduced genital lesions and asymptomatic HSV-2 genital shedding in guinea pigs, yet the protection was not complete. We evaluated whether adding the T cell immunogens UL19 (capsid protein VP5) and UL47 (tegument protein VP13/14) would enhance the protection provided by the gC2/gD2 vaccine, which produces potent antibody responses. Here we report the efficacy of a combination vaccine containing gC2/gD2 and UL19/UL47 for prevention of genital disease, vaginal shedding of HSV-2 DNA, and latent infection of dorsal root ganglia in guinea pigs.
Collapse
|
23
|
Quinn KM, Zak DE, Costa A, Yamamoto A, Kastenmuller K, Hill BJ, Lynn GM, Darrah PA, Lindsay RWB, Wang L, Cheng C, Nicosia A, Folgori A, Colloca S, Cortese R, Gostick E, Price DA, Gall JGD, Roederer M, Aderem A, Seder RA. Antigen expression determines adenoviral vaccine potency independent of IFN and STING signaling. J Clin Invest 2015; 125:1129-46. [PMID: 25642773 DOI: 10.1172/jci78280] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 12/23/2014] [Indexed: 12/13/2022] Open
Abstract
Recombinant adenoviral vectors (rAds) are lead vaccine candidates for protection against a variety of pathogens, including Ebola, HIV, tuberculosis, and malaria, due to their ability to potently induce T cell immunity in humans. However, the ability to induce protective cellular immunity varies among rAds. Here, we assessed the mechanisms that control the potency of CD8 T cell responses in murine models following vaccination with human-, chimpanzee-, and simian-derived rAds encoding SIV-Gag antigen (Ag). After rAd vaccination, we quantified Ag expression and performed expression profiling of innate immune response genes in the draining lymph node. Human-derived rAd5 and chimpanzee-derived chAd3 were the most potent rAds and induced high and persistent Ag expression with low innate gene activation, while less potent rAds induced less Ag expression and robustly induced innate immunity genes that were primarily associated with IFN signaling. Abrogation of type I IFN or stimulator of IFN genes (STING) signaling increased Ag expression and accelerated CD8 T cell response kinetics but did not alter memory responses or protection. These findings reveal that the magnitude of rAd-induced memory CD8 T cell immune responses correlates with Ag expression but is independent of IFN and STING and provide criteria for optimizing protective CD8 T cell immunity with rAd vaccines.
Collapse
|
24
|
Majhen D, Calderon H, Chandra N, Fajardo CA, Rajan A, Alemany R, Custers J. Adenovirus-based vaccines for fighting infectious diseases and cancer: progress in the field. Hum Gene Ther 2014; 25:301-17. [PMID: 24580050 DOI: 10.1089/hum.2013.235] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
The field of adenovirology is undergoing rapid change in response to increasing appreciation of the potential advantages of adenoviruses as the basis for new vaccines and as vectors for gene and cancer therapy. Substantial knowledge and understanding of adenoviruses at a molecular level has made their manipulation for use as vaccines and therapeutics relatively straightforward in comparison with other viral vectors. In this review we summarize the structure and life cycle of the adenovirus and focus on the use of adenovirus-based vectors in vaccines against infectious diseases and cancers. Strategies to overcome the problem of preexisting antiadenovirus immunity, which can hamper the immunogenicity of adenovirus-based vaccines, are discussed. When armed with tumor-associated antigens, replication-deficient and oncolytic adenoviruses can efficiently activate an antitumor immune response. We present concepts on how to use adenoviruses as therapeutic cancer vaccines and consider some of the strategies used to further improve antitumor immune responses. Studies that explore the prospect of adenoviruses as vaccines against infectious diseases and cancer are underway, and here we give an overview of the latest developments.
Collapse
|
25
|
Johnson MJ, Björkström NK, Petrovas C, Liang F, Gall JGD, Loré K, Koup RA. Type I interferon-dependent activation of NK cells by rAd28 or rAd35, but not rAd5, leads to loss of vector-insert expression. Vaccine 2013; 32:717-24. [PMID: 24325826 DOI: 10.1016/j.vaccine.2013.11.055] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Revised: 11/08/2013] [Accepted: 11/15/2013] [Indexed: 01/05/2023]
Abstract
Vaccines constructed from rare-serotype recombinant adenovirus vectors (rAd) such as rAd serotype 28 (rAd28) and rAd35 are currently being explored as alternatives to rAd5-based vaccines because they circumvent the problems with pre-existing immunity that complicate the effectiveness of rAd5 vaccines. However, previous work has demonstrated that the immunogenicity of rAd28 and rAd35 is substantially lower than rAd5. Here we show that rAd28 and rAd35 increase apoptosis of antigen presenting cells (APCs), such as monocytes, relative to rAd5 and mock infected controls. APCs undergoing apoptosis showed an increased loss of vector-insert expression. Loss of vector-insert expression correlated with activation of NK cells, which resulted in apoptosis of co-cultured monocytes. Finally, we show that activation of NK cells is dependent on IFNα which is produced by exposure to rAd28 or rAd35, but not to rAd5. Taken together, these data demonstrate that IFNα-induced activation of NK cells leads to increased monocyte apoptosis and subsequent vector-insert loss. This may be a possible mechanism that results in reduced immunogenicity of rAd28 and rAd35-based vectors.
Collapse
Affiliation(s)
- Matthew J Johnson
- Immunology Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA; Department of Microbiology and Immunology, Georgetown University Medical Center, Washington, DC 20007, USA
| | - Niklas K Björkström
- Pathology Department, Center for Infectious Medicine, Karolinska Institutet, Stockholm 14186, Sweden; Department of Medicine, Karolinska Institutet, Stockholm 14186, Sweden
| | - Constantinos Petrovas
- Immunology Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Frank Liang
- Immunology Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA; Pathology Department, Center for Infectious Medicine, Karolinska Institutet, Stockholm 14186, Sweden; Department of Medicine, Karolinska Institutet, Stockholm 14186, Sweden
| | | | - Karin Loré
- Immunology Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA; Pathology Department, Center for Infectious Medicine, Karolinska Institutet, Stockholm 14186, Sweden; Department of Medicine, Karolinska Institutet, Stockholm 14186, Sweden
| | - Richard A Koup
- Immunology Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
26
|
Duncan M, Cranfield MR, Torano H, Kuete HM, Lee GP, Glenn A, Bruder JT, Rangel D, Brough DE, Gall JG. Adenoviruses isolated from wild gorillas are closely related to human species C viruses. Virology 2013; 444:119-23. [PMID: 23806387 DOI: 10.1016/j.virol.2013.05.041] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Revised: 05/30/2013] [Accepted: 05/31/2013] [Indexed: 11/26/2022]
Abstract
We have isolated and cultured three distinct adenoviruses from wild gorillas. Phylogenetic analysis grouped the viruses with human adenovirus species C based on DNA polymerase, hexon, and E4ORF6 genes. The three wild gorilla adenoviruses clustered with the other species C captive gorilla adenoviruses, forming a branch separate from human and chimpanzee/bonobo adenoviruses. Animal sera to the three newly isolated viruses did not cross-neutralize, demonstrating serological distinctiveness. The human adenovirus 5 fiber knob blocked infection, suggesting use of the Coxsackie and Adenovirus Receptor. These viruses may provide viral vectors with properties distinct from chimpanzee adenovirus and human adenovirus vectors.
Collapse
Affiliation(s)
- McVey Duncan
- GenVec, Inc., 65W. Watkins Mill Rd, Gaithersburg, MD 20878, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Johnson TR, Rangel D, Graham BS, Brough DE, Gall JG. Genetic vaccine for respiratory syncytial virus provides protection without disease potentiation. Mol Ther 2013; 22:196-205. [PMID: 23752342 DOI: 10.1038/mt.2013.142] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Accepted: 06/03/2013] [Indexed: 01/05/2023] Open
Abstract
Respiratory syncytial virus (RSV) is a major cause of infectious lower respiratory disease in infants and the elderly. As there is no vaccine for RSV, we developed a genetic vaccine approach that induced protection of the entire respiratory tract from a single parenteral administration. The approach was based on adenovirus vectors derived from newly isolated nonhuman primate viruses with low seroprevalence. We show for the first time that a single intramuscular (IM) injection of the replication-deficient adenovirus vectors expressing the RSV fusion (F0) glycoprotein induced immune responses that protected both the lungs and noses of cotton rats and mice even at low doses and for several months postimmunization. The immune response included high titers of neutralizing antibody that were maintained ≥ 24 weeks and RSV-specific CD8+ and CD4+ T cells. The vectors were as potently immunogenic as a human adenovirus 5 vector in these two key respiratory pathogen animal models. Importantly, there was minimal alveolitis and granulocytic infiltrates in the lung, and type 2 cytokines were not produced after RSV challenge even under conditions of partial protection. Overall, this genetic vaccine is highly effective without potentiating immunopathology, and the results support development of the vaccine candidate for human testing.
Collapse
Affiliation(s)
| | | | - Barney S Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | | | | |
Collapse
|
28
|
Prospects for oral replicating adenovirus-vectored vaccines. Vaccine 2013; 31:3236-43. [PMID: 23707160 DOI: 10.1016/j.vaccine.2013.05.016] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Revised: 05/06/2013] [Accepted: 05/07/2013] [Indexed: 12/22/2022]
Abstract
Orally delivered replicating adenovirus (Ad) vaccines have been used for decades to prevent adenovirus serotype 4 and 7 respiratory illness in military recruits, demonstrating exemplary safety and high efficacy. That experience suggests that oral administration of live recombinant Ads (rAds) holds promise for immunization against other infectious diseases, including those that have been refractory to traditional vaccination methods. Live rAds can express intact antigens from free-standing transgenes during replication in infected cells. Alternatively, antigenic epitopes can be displayed on the rAd capsid itself, allowing presentation of the epitope to the immune system both prior to and during replication of the virus. Such capsid-display rAds offer a novel vaccine approach that could be used either independently of or in combination with transgene expression strategies to provide a new tool in the search for protection from infectious disease.
Collapse
|
29
|
Wei CJ, Yassine HM, McTamney PM, Gall JGD, Whittle JRR, Boyington JC, Nabel GJ. Elicitation of broadly neutralizing influenza antibodies in animals with previous influenza exposure. Sci Transl Med 2013; 4:147ra114. [PMID: 22896678 DOI: 10.1126/scitranslmed.3004273] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
The immune system responds to influenza infection by producing neutralizing antibodies to the viral surface protein, hemagglutinin (HA), which regularly changes its antigenic structure. Antibodies that target the highly conserved stem region of HA neutralize diverse influenza viruses and can be elicited through vaccination in animals and humans. Efforts to develop universal influenza vaccines have focused on strategies to elicit such antibodies; however, the concern has been raised that previous influenza immunity may abrogate the induction of such broadly protective antibodies. We show here that prime-boost immunization can induce broadly neutralizing antibody responses in influenza-immune mice and ferrets that were previously infected or vaccinated. HA stem-directed antibodies were elicited in mice primed with a DNA vaccine and boosted with inactivated vaccine from H1N1 A/New Caledonia/20/1999 (1999 NC) HA regardless of preexposure. Similarly, gene-based vaccination with replication-defective adenovirus 28 (rAd28) and 5 (rAd5) vectors encoding 1999 NC HA elicited stem-directed neutralizing antibodies and conferred protection against unmatched 1934 and 2007 H1N1 virus challenge in influenza-immune ferrets. Indeed, previous exposure to certain strains could enhance immunogenicity: The strongest HA stem-directed immune response was observed in ferrets previously infected with a divergent 1934 H1N1 virus. These findings suggest that broadly neutralizing antibodies against the conserved stem region of HA can be elicited through vaccination despite previous influenza exposure, which supports the feasibility of developing stem-directed universal influenza vaccines for humans.
Collapse
Affiliation(s)
- Chih-Jen Wei
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892-3005, USA
| | | | | | | | | | | | | |
Collapse
|
30
|
Quinn KM, Da Costa A, Yamamoto A, Berry D, Lindsay RWB, Darrah PA, Wang L, Cheng C, Kong WP, Gall JGD, Nicosia A, Folgori A, Colloca S, Cortese R, Gostick E, Price DA, Gomez CE, Esteban M, Wyatt LS, Moss B, Morgan C, Roederer M, Bailer RT, Nabel GJ, Koup RA, Seder RA. Comparative analysis of the magnitude, quality, phenotype, and protective capacity of simian immunodeficiency virus gag-specific CD8+ T cells following human-, simian-, and chimpanzee-derived recombinant adenoviral vector immunization. THE JOURNAL OF IMMUNOLOGY 2013; 190:2720-35. [PMID: 23390298 DOI: 10.4049/jimmunol.1202861] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Recombinant adenoviral vectors (rAds) are the most potent recombinant vaccines for eliciting CD8(+) T cell-mediated immunity in humans; however, prior exposure from natural adenoviral infection can decrease such responses. In this study we show low seroreactivity in humans against simian- (sAd11, sAd16) or chimpanzee-derived (chAd3, chAd63) compared with human-derived (rAd5, rAd28, rAd35) vectors across multiple geographic regions. We then compared the magnitude, quality, phenotype, and protective capacity of CD8(+) T cell responses in mice vaccinated with rAds encoding SIV Gag. Using a dose range (1 × 10(7)-10(9) particle units), we defined a hierarchy among rAd vectors based on the magnitude and protective capacity of CD8(+) T cell responses, from most to least, as: rAd5 and chAd3, rAd28 and sAd11, chAd63, sAd16, and rAd35. Selection of rAd vector or dose could modulate the proportion and/or frequency of IFN-γ(+)TNF-α(+)IL-2(+) and KLRG1(+)CD127(-)CD8(+) T cells, but strikingly ∼30-80% of memory CD8(+) T cells coexpressed CD127 and KLRG1. To further optimize CD8(+) T cell responses, we assessed rAds as part of prime-boost regimens. Mice primed with rAds and boosted with NYVAC generated Gag-specific responses that approached ∼60% of total CD8(+) T cells at peak. Alternatively, priming with DNA or rAd28 and boosting with rAd5 or chAd3 induced robust and equivalent CD8(+) T cell responses compared with prime or boost alone. Collectively, these data provide the immunologic basis for using specific rAd vectors alone or as part of prime-boost regimens to induce CD8(+) T cells for rapid effector function or robust long-term memory, respectively.
Collapse
Affiliation(s)
- Kylie M Quinn
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Immunogenicity of a recombinant measles-HIV-1 clade B candidate vaccine. PLoS One 2012; 7:e50397. [PMID: 23226275 PMCID: PMC3511521 DOI: 10.1371/journal.pone.0050397] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2012] [Accepted: 10/19/2012] [Indexed: 01/08/2023] Open
Abstract
Live attenuated measles virus is one of the most efficient and safest vaccines available, making it an attractive candidate vector for a HIV/AIDS vaccine aimed at eliciting cell-mediated immune responses (CMI). Here we have characterized the potency of CMI responses generated in mice and non-human primates after intramuscular immunisation with a candidate recombinant measles vaccine carrying an HIV-1 insert encoding Clade B Gag, RT and Nef (MV1-F4). Eight Mauritian derived, MHC-typed cynomolgus macaques were immunised with 105 TCID50 of MV1-F4, four of which were boosted 28 days later with the same vaccine. F4 and measles virus (MV)-specific cytokine producing T cell responses were detected in 6 and 7 out of 8 vaccinees, respectively. Vaccinees with either M6 or recombinant MHC haplotypes demonstrated the strongest cytokine responses to F4 peptides. Polyfunctional analysis revealed a pattern of TNFα and IL-2 responses by CD4+ T cells and TNFα and IFNγ responses by CD8+ T cells to F4 peptides. HIV-specific CD4+ and CD8+ T cells expressing cytokines waned in peripheral blood lymphocytes by day 84, but CD8+ T cell responses to F4 peptides could still be detected in lymphoid tissues more than 3 months after vaccination. Anti-F4 and anti-MV antibody responses were detected in 6 and 8 out of 8 vaccinees, respectively. Titres of anti-F4 and MV antibodies were boosted in vaccinees that received a second immunisation. MV1-F4 carrying HIV-1 Clade B inserts induces robust boostable immunity in non-human primates. These results support further exploration of the MV1-F4 vector modality in vaccination strategies that may limit HIV-1 infectivity.
Collapse
|
32
|
Saxena M, Van TTH, Baird FJ, Coloe PJ, Smooker PM. Pre-existing immunity against vaccine vectors--friend or foe? MICROBIOLOGY-SGM 2012; 159:1-11. [PMID: 23175507 PMCID: PMC3542731 DOI: 10.1099/mic.0.049601-0] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Over the last century, the successful attenuation of multiple bacterial and viral pathogens has led to an effective, robust and safe form of vaccination. Recently, these vaccines have been evaluated as delivery vectors for heterologous antigens, as a means of simultaneous vaccination against two pathogens. The general consensus from published studies is that these vaccine vectors have the potential to be both safe and efficacious. However, some of the commonly employed vectors, for example Salmonella and adenovirus, often have pre-existing immune responses in the host and this has the potential to modify the subsequent immune response to a vectored antigen. This review examines the literature on this topic, and concludes that for bacterial vectors there can in fact, in some cases, be an enhancement in immunogenicity, typically humoral, while for viral vectors pre-existing immunity is a hindrance for subsequent induction of cell-mediated responses.
Collapse
Affiliation(s)
- Manvendra Saxena
- Ludwig Institute for Cancer Research, Heidelberg, Victoria, Australia
| | - Thi Thu Hao Van
- School of Applied Sciences, RMIT University, Bundoora, Victoria, Australia
| | - Fiona J Baird
- Comparative Genomics Centre, School of Pharmacy and Molecular Sciences, James Cook University, Townsville, Queensland, Australia
| | - Peter J Coloe
- School of Applied Sciences, RMIT University, Bundoora, Victoria, Australia
| | - Peter M Smooker
- School of Applied Sciences, RMIT University, Bundoora, Victoria, Australia
| |
Collapse
|
33
|
Abstract
The emergence of a highly pathogenic avian influenza virus H5N1 has increased the potential for a new pandemic to occur. This event highlights the necessity for developing a new generation of influenza vaccines to counteract influenza disease. These vaccines must be manufactured for mass immunization of humans in a timely manner. Poultry should be included in this policy, since persistent infected flocks are the major source of avian influenza for human infections. Recombinant adenoviral vectored H5N1 vaccines are an attractive alternative to the currently licensed influenza vaccines. This class of vaccines induces a broadly protective immunity against antigenically distinct H5N1, can be manufactured rapidly, and may allow mass immunization of human and poultry. Recombinant adenoviral vectors derived from both human and non-human adenoviruses are currently being investigated and appear promising both in nonclinical and clinical studies. This review will highlight the current status of various adenoviral vectored H5N1 vaccines and will outline novel approaches for the future.
Collapse
|
34
|
Abstract
Viral vectors have been developed as vaccine platforms for a number of pathogens and tumors. In particular, adenovirus (Ad)-based vectors expressing genes coding for pathogen or tumor antigens have proven efficacious to induce protective immunity. Major challenges in the use of Ad vectors are the high prevalence of anti-Ad immunity and the recent observation during an Ad-based HIV vaccine trial that led to increased HIV-1 acquisition in the presence of circulating anti-Ad5 neutralizing antibodies. In this review we summarize strategies to address these challenges and focus on modifications of the Ad capsid to enhance the adjuvant effect of anti-Ad immunogenicity and to circumvent pre-existing immunity. In addition, we summarize the current status and potential of other viral vector vaccines based on adeno-associated viruses, lentiviruses and poxviruses.
Collapse
|
35
|
Bolhassani A, Zahedifard F. Therapeutic live vaccines as a potential anticancer strategy. Int J Cancer 2012; 131:1733-43. [PMID: 22610886 DOI: 10.1002/ijc.27640] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2012] [Accepted: 05/10/2012] [Indexed: 01/13/2023]
Abstract
The design of efficient cancer treatments is one of the major challenges of medical science. Therapeutic vaccines of cancer have been emerged as an attractive approach for their capacity of breaking the immune tolerance and invoking long-term immune response targeting cancer cells without autoimmunity. An efficient antigen delivery system is the key issue of developing an effective cancer vaccine. In this regard, live vaccination strategies including various live bacterial and viral vectors have attracted a great attention. Several bacterial strains such as Salmonella, Listeria monocytogenes and Lactococcus lactis effectively colonize solid tumors and act as antitumor therapeutics. On the other hand, the use of viruses as vaccine vectors such as Vaccinia, Adenovirus, Herpes simplex virus, Paramyxovirus and Retroviruses utilizes mechanisms that evolved in these microbes for entering cells and capturing the cellular machinery to express viral proteins. Viral/bacterial-vectored vaccines induce systemic T-cell responses including polyfunctional cytokine-secreting CD4+ and CD8+ T-cells. However, there is an urgent need for the development of new safe live vaccine vectors that are capable of enhancing antigen presentation and eliciting potent immune responses without the risk of development of disease in humans. Recently, nonpathogenic parasites including Leishmania tarentolae, Toxoplasma gondii and Trypanosoma cruzi have emerged to be a novel candidate for gene delivery and heterologous genes expression. In this review, recent researches on cancer therapy using genetically modified bacteria and virus are summarized. In addition, live parasite-based vectors will be discussed as a novel anticancer therapeutic approach.
Collapse
Affiliation(s)
- Azam Bolhassani
- Molecular Immunology and Vaccine Research Laboratory, Pasteur Institute of Iran, Tehran, Iran.
| | | |
Collapse
|
36
|
Johnson MJ, Petrovas C, Yamamoto T, Lindsay RWB, Loré K, Gall JGD, Gostick E, Lefebvre F, Cameron MJ, Price DA, Haddad E, Sekaly RP, Seder RA, Koup RA. Type I IFN induced by adenovirus serotypes 28 and 35 has multiple effects on T cell immunogenicity. THE JOURNAL OF IMMUNOLOGY 2012; 188:6109-18. [PMID: 22586038 DOI: 10.4049/jimmunol.1103717] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Recombinant adenovirus (rAd) vectors are being investigated as vaccine delivery vehicles in preclinical and clinical studies. rAds constructed from different serotypes differ in receptor usage, tropism, and ability to activate cells, aspects of which likely contribute to their different immunogenicity profiles. In this study, we compared the infectivity and cell stimulatory capacity of recombinant adenovirus serotype 5 (rAd5), recombinant adenovirus serotype 28 (rAd28), and recombinant adenovirus serotype 35 (rAd35) in association with their respective immunogenicity profiles. We found that rAd28 and rAd35 infected and led to the in vitro maturation and activation of both human and mouse dendritic cells more efficiently compared with rAd5. In stark contrast to rAd5, rAd28 and rAd35 induced production of IFN-α and stimulated IFN-related intracellular pathways. However, the in vivo immunogenicity of rAd28 and rAd35 was significantly lower than that of rAd5. Deletion of IFN-α signaling during vaccination with rAd28 and rAd35 vectors increased the magnitude of the insert-specific T cell response to levels induced by vaccination with rAd5 vector. The negative impact of IFN-α signaling on the magnitude of the T cell response could be overcome by increasing the vaccine dose, which was also associated with greater polyfunctionality and a more favorable long-term memory phenotype of the CD8 T cell response in the presence of IFN-α signaling. Taken together, our results demonstrate that rAd-induced IFN-α production has multiple effects on T cell immunogenicity, the understanding of which should be considered in the design of rAd vaccine vectors.
Collapse
Affiliation(s)
- Matthew J Johnson
- Immunology Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Engineered Viruses as Vaccine Platforms. INNOVATION IN VACCINOLOGY 2012. [PMCID: PMC7120934 DOI: 10.1007/978-94-007-4543-8_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Many viruses have been investigated for the development of genetic vaccines and the ideal ones must be endowed with many properties, such as the quality and the quantity of the immunological response induced against the encoded antigens, safety and production on a large scale basis. Viral based vaccines must also deal with the potential problem of the pre-existing antivector immunity. Several viral vaccine vectors have emerged to date, all of them having relative advantages and limits depending on the proposed application. Recent successes reflect diverse improvements such as development of new adenovirus serotypes and prime-boost regimes. This chapter describes the features of four viral vector systems based on poxviruses, adenoviruses, alphaviruses and lentiviruses and recent results following their use with a particular emphasis on clinical research, highlighting the challenges and successes.
Collapse
|
38
|
Abstract
Progress in vector design and an increased knowledge of mechanisms underlying tumor-induced immune suppression have led to a new and promising generation of Adenovirus (Ad)-based immunotherapies, which are discussed in this review. As vaccine vehicles Ad vectors (AdVs) have been clinically evaluated and proven safe, but a major limitation of the commonly used Ad5 serotype is neutralization by preexistent or rapidly induced immune responses. Genetic modifications in the Ad capsid can reduce intrinsic immunogenicity and facilitate escape from antibody-mediated neutralization. Further modification of the Ad hexon and fiber allows for liver and scavenger detargeting and selective targeting of, for example, dendritic cells. These next-generation Ad vaccines with enhanced efficacy are now becoming available for testing as tumor vaccines. In addition, AdVs encoding immune-modulating products may be used to convert the tumor microenvironment from immune-suppressive and proinvasive to proinflammatory, thus facilitating cell-mediated effector functions that can keep tumor growth and invasion in check. Oncolytic AdVs, that selectively replicate in tumor cells and induce an immunogenic form of cell death, can also be armed with immune-activating transgenes to amplify primed antitumor immune responses. These novel immunotherapy strategies, employing highly efficacious AdVs in optimized configurations, show great promise and warrant clinical exploration.
Collapse
|
39
|
Schlecker C, Praetorius M, Brough DE, Presler RG, Hsu C, Plinkert PK, Staecker H. Selective atonal gene delivery improves balance function in a mouse model of vestibular disease. Gene Ther 2011; 18:884-90. [PMID: 21472006 PMCID: PMC3136627 DOI: 10.1038/gt.2011.33] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2010] [Revised: 11/22/2010] [Accepted: 11/22/2010] [Indexed: 11/08/2022]
Abstract
Loss of balance is often due to loss of vestibular hair cells. In mammals, regeneration of functional hair cells in the mature sensory epithelium is limited; therefore, loss of sensory cells can lead to debilitating balance problems. Delivery of the transcription factor atonal (atoh1) after aminoglycoside ototoxicity has previously been shown to induce the transdifferentiation of supporting cells into new hair cells and restore function. A problem with mouse aminoglycoside models is that the partial loss of hair cells seen in human disease is difficult to establish consistently. To more closely mirror human clinical balance dysfunction, we have used systemic application of 3,3'-iminodipropionitrile (IDPN), a vestibulotoxic nitrile compound known to cause vestibular hair cell loss, to induce a consistent partial loss of vestibular hair cells. To determine if balance function could be restored, we delivered atoh1 using a new adenovirus vector, based on Ad28. The Ad28 adenovector is based on a human serotype with a low seroprevalence that appears to target gene delivery to vestibular supporting cells. To further provide cell type selectivity of gene delivery, we expressed atoh1 using the supporting cell-specific glial fibrillary acid protein promoter. Delivery of this vector to IDPN-damaged vestibular organs resulted in a significant recovery of vestibular hair cells and restoration of balance, as measured by time on rotarod compared with untreated controls.
Collapse
Affiliation(s)
- Christina Schlecker
- Department of Otolaryngology, Head and Neck Surgery, University of Kansas School of Medicine, Kansas City, Kansas, USA
- Department of Otolaryngology, University of Heidelberg, Heidelberg, Germany
| | - Mark Praetorius
- Department of Otolaryngology, Head and Neck Surgery, University of Kansas School of Medicine, Kansas City, Kansas, USA
- Department of Otolaryngology, University of Heidelberg, Heidelberg, Germany
| | | | | | - Chi Hsu
- GenVec Inc., Gaithersburg, Maryland, USA
| | - Peter K. Plinkert
- Department of Otolaryngology, University of Heidelberg, Heidelberg, Germany
| | - Hinrich Staecker
- Department of Otolaryngology, Head and Neck Surgery, University of Kansas School of Medicine, Kansas City, Kansas, USA
| |
Collapse
|
40
|
Rollier CS, Reyes-Sandoval A, Cottingham MG, Ewer K, Hill AVS. Viral vectors as vaccine platforms: deployment in sight. Curr Opin Immunol 2011; 23:377-82. [DOI: 10.1016/j.coi.2011.03.006] [Citation(s) in RCA: 144] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2011] [Accepted: 03/25/2011] [Indexed: 02/06/2023]
|
41
|
"In vitro systems to characterize the immune response to HIV-1 and HIV-1 vaccine candidates", NIAID Workshop Report, Bethesda, August 4, 2010. Vaccine 2011; 29:4647-53. [PMID: 21571022 DOI: 10.1016/j.vaccine.2011.04.035] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2011] [Revised: 04/01/2011] [Accepted: 04/12/2011] [Indexed: 11/23/2022]
Abstract
Although clinical trials are the ultimate way to prove vaccine safety and efficacy, the complexity, cost and time required to develop a product to enter human trials demand a serious, long-term investment. Lack of knowledge on immune correlates of protection from HIV infections makes investments in HIV vaccine research significantly risky. Preclinical testing of HIV vaccines is routinely carried out in non-human primate models however these studies have a significant cost and their predictive value is still questionable. The potential value of screening new HIV-1 vaccine candidates on human cells and tissues via high throughput in vitro systems that allow rapid, cost-effective and accurate predictions of in vivo immune responses would be enormous. A one-day workshop was convened by Division of AIDS, National Institutes of Health on August 4, 2010 to address the benefits and challenges of assessing HIV-1 vaccine responses in alternative ways. Consideration was given to the use of various in vitro model systems, human mucosal tissue explants and humanized mouse models as ways to predict immunogenicity and efficacy of HIV-1 vaccines early in the development process, and support decisions on whether a product may be worthy of moving into non-human primates or human trials. This report summarizes the outcome of the workshop.
Collapse
|
42
|
Robinson CM, Seto D, Jones MS, Dyer DW, Chodosh J. Molecular evolution of human species D adenoviruses. INFECTION GENETICS AND EVOLUTION 2011; 11:1208-17. [PMID: 21570490 DOI: 10.1016/j.meegid.2011.04.031] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2011] [Revised: 04/26/2011] [Accepted: 04/27/2011] [Indexed: 12/15/2022]
Abstract
Adenoviruses are medium-sized double stranded DNA viruses that infect vertebrates. Human adenoviruses cause an array of diseases. Currently there are 56 human adenovirus types recognized and characterized within seven species (A-G). Of those types, a majority belongs to species D. In this review, the genomic conservation and diversity are examined among human adenoviruses within species D, particularly in contrast to other human adenovirus species. Specifically, homologous recombination is presented as a driving force for the molecular evolution of human adenoviruses and the emergence of new adenovirus pathogens.
Collapse
Affiliation(s)
- Christopher M Robinson
- Howe Laboratory, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, 243 Charles Street, Boston, MA 02114, USA. USA
| | | | | | | | | |
Collapse
|